551
|
Slepicka PF, Cyrill SL, Dos Santos CO. Pregnancy and Breast Cancer: Pathways to Understand Risk and Prevention. Trends Mol Med 2019; 25:866-881. [PMID: 31383623 DOI: 10.1016/j.molmed.2019.06.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
Several studies have made strong efforts to understand how age and parity modulate the risk of breast cancer. A holistic understanding of the dynamic regulation of the morphological, cellular, and molecular milieu of the mammary gland offers insights into the drivers of breast cancer development as well as into potential prophylactic interventions, the latter being a longstanding ambition of the research and clinical community aspiring to eradicate the disease. In this review we discuss mechanisms that react to pregnancy signals, and we delineate the nuances of pregnancy-associated dynamism that contribute towards either breast cancer development or prevention. Further definition of the molecular basis of parity and breast cancer risk may allow the elaboration of tools to predict and survey those who are at risk of breast cancer development.
Collapse
Affiliation(s)
- Priscila F Slepicka
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Samantha L Cyrill
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
552
|
Abstract
Triple-negative breast cancer (TNBC) is a breast cancer subtype renowned for its capacity to affect younger women, metastasise early despite optimal adjuvant treatment and carry a poor prognosis. Neoadjuvant therapy has focused on combinations of systemic agents to optimise pathological complete response. Treatment algorithms now guide the management of patients with or without residual disease, but metastatic TNBC continues to harbour a poor prognosis. Innovative, multi-drug combination systemic therapies in the neoadjuvant and adjuvant settings have led to significant improvements in outcomes, particularly over the past decade. Recently published advances in the treatment of metastatic TNBC have shown impressive results with poly (ADP-ribose) polymerase (PARP) inhibitors and immunotherapy agents. Immunotherapy agents in combination with traditional systemic chemotherapy have been shown to alter the natural history of this devastating condition, particularly in patients whose tumours are positive for programmed cell death ligand 1 (PD-L1).
Collapse
Affiliation(s)
| | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
553
|
Waks AG, Stover DG, Guerriero JL, Dillon D, Barry WT, Gjini E, Hartl C, Lo W, Savoie J, Brock J, Wesolowski R, Li Z, Damicis A, Philips AV, Wu Y, Yang F, Sullivan A, Danaher P, Brauer HA, Osmani W, Lipschitz M, Hoadley KA, Goldberg M, Perou CM, Rodig S, Winer EP, Krop IE, Mittendorf EA, Tolaney SM. The Immune Microenvironment in Hormone Receptor-Positive Breast Cancer Before and After Preoperative Chemotherapy. Clin Cancer Res 2019; 25:4644-4655. [PMID: 31061067 PMCID: PMC6677598 DOI: 10.1158/1078-0432.ccr-19-0173] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/05/2019] [Accepted: 05/01/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Hormone receptor-positive/HER2-negative (HR+/HER2-) breast cancer is associated with low levels of stromal tumor-infiltrating lymphocytes (sTIL) and PD-L1, and demonstrates poor responses to checkpoint inhibitor therapy. Evaluating the effect of standard chemotherapy on the immune microenvironment may suggest new opportunities for immunotherapy-based approaches to treating HR+/HER2- breast tumors. EXPERIMENTAL DESIGN HR+/HER2- breast tumors were analyzed before and after neoadjuvant chemotherapy. sTIL were assessed histologically; CD8+ cells, CD68+ cells, and PD-L1 staining were assessed immunohistochemically; whole transcriptome sequencing and panel RNA expression analysis (NanoString) were performed. RESULTS Ninety-six patients were analyzed from two cohorts (n = 55, Dana-Farber cohort; n = 41, MD Anderson cohort). sTIL, CD8, and PD-L1 on tumor cells were higher in tumors with basal PAM50 intrinsic subtype. Higher levels of tissue-based lymphocyte (sTIL, CD8, PD-L1) and macrophage (CD68) markers, as well as gene expression markers of lymphocyte or macrophage phenotypes (NanoString or CIBERSORT), correlated with favorable response to neoadjuvant chemotherapy, but not with improved distant metastasis-free survival in these cohorts or a large gene expression dataset (N = 302). In paired pre-/postchemotherapy samples, sTIL and CD8+ cells were significantly decreased after treatment, whereas expression analyses (NanoString) demonstrated significant increase of multiple myeloid signatures. Single gene expression implicated increased expression of immunosuppressive (M2-like) macrophage-specific genes after chemotherapy. CONCLUSIONS The immune microenvironment of HR+/HER2- tumors differs according to tumor biology. This cohort of paired pre-/postchemotherapy samples suggests a critical role for immunosuppressive macrophage expansion in residual disease. The role of macrophages in chemoresistance should be explored, and further evaluation of macrophage-targeting therapy is warranted.
Collapse
Affiliation(s)
- Adrienne G Waks
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Daniel G Stover
- Division of Medical Oncology, Ohio State University College of Medicine, Columbus, Ohio
| | - Jennifer L Guerriero
- Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - William T Barry
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Evisa Gjini
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Christina Hartl
- Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wesley Lo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer Savoie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jane Brock
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robert Wesolowski
- Division of Medical Oncology, Ohio State University College of Medicine, Columbus, Ohio
| | - Zaibo Li
- Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio
| | - Adrienne Damicis
- Department of Biostatistics, Ohio State University College of Public Health, Columbus, Ohio
| | - Anne V Philips
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yun Wu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fei Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Wafa Osmani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mikel Lipschitz
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Katherine A Hoadley
- University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Michael Goldberg
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles M Perou
- University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Eric P Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ian E Krop
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elizabeth A Mittendorf
- Breast Tumor Immunology Laboratory, Susan F. Smith Center for Women's Cancers, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
554
|
Vinayak S, Tolaney SM, Schwartzberg L, Mita M, McCann G, Tan AR, Wahner-Hendrickson AE, Forero A, Anders C, Wulf GM, Dillon P, Lynce F, Zarwan C, Erban JK, Zhou Y, Buerstatte N, Graham JR, Arora S, Dezube BJ, Telli ML. Open-label Clinical Trial of Niraparib Combined With Pembrolizumab for Treatment of Advanced or Metastatic Triple-Negative Breast Cancer. JAMA Oncol 2019; 5:1132-1140. [PMID: 31194225 DOI: 10.1001/jamaoncol.2019.1029] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Importance Poly(adenosine diphosphate-ribose) polymerase inhibitor and anti-programmed death receptor-1 inhibitor monotherapy have shown limited clinical activity in patients with advanced triple-negative breast cancer (TNBC). Objective To evaluate the clinical activity (primary) and safety (secondary) of combination treatment with niraparib and pembrolizumab in patients with advanced or metastatic TNBC. Design, Setting, and Participants This open-label, single-arm, phase 2 study enrolled 55 eligible patients with advanced or metastatic TNBC irrespective of BRCA mutation status or programmed death-ligand 1 (PD-L1) expression at 34 US sites. Data were collected from January 3, 2017, through October 29, 2018, and analyzed from October 29, 2018, through February 27, 2019. Interventions Patients were administered 200 mg of oral niraparib once daily in combination with 200 mg of intravenous pembrolizumab on day 1 of each 21-day cycle. Main Outcomes and Measures The primary end point was objective response rate (ORR) per the Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary end points were safety, disease control rate (DCR; complete response plus partial response plus stable disease), duration of response (DOR), progression-free survival (PFS), and overall survival. Results Within the full study population of 55 women (median age, 54 years [range, 32-90 years]), 5 patients had confirmed complete responses, 5 had confirmed partial responses, 13 had stable disease, and 24 had progressive disease. In the efficacy-evaluable population (n = 47), ORR included 10 patients (21%; 90% CI, 12%-33%) and DCR included 23 (49%; 90% CI, 36%-62%). Median DOR was not reached at the time of the data cutoff, with 7 patients still receiving treatment at the time of analysis. In 15 evaluable patients with tumor BRCA mutations, ORR included 7 patients(47%; 90% CI, 24%-70%), DCR included 12 (80%; 90% CI, 56%-94%), and median PFS was 8.3 months (95% CI, 2.1 months to not estimable). In 27 evaluable patients with BRCA wild-type tumors, ORR included 3 patients (11%; 90% CI, 3%-26%), DCR included 9 (33%; 90% CI, 19%-51%), and median PFS was 2.1 months (95% CI, 1.4-2.5 months). The most common treatment-related adverse events of grade 3 or higher were anemia (10 [18%]), thrombocytopenia (8 [15%]), and fatigue (4 [7%]). Immune-related adverse events were reported in 8 patients (15%) and were grade 3 in 2 patients (4%); no new safety signals were detected. Conclusions and Relevance Combination niraparib plus pembrolizumab provides promising antitumor activity in patients with advanced or metastatic TNBC, with numerically higher response rates in those with tumor BRCA mutations. The combination therapy was safe with a tolerable safety profile, warranting further investigation. Trial Registration ClinicalTrials.gov identifier: NCT02657889.
Collapse
Affiliation(s)
- Shaveta Vinayak
- Case Comprehensive Cancer Center, University Hospitals, Case Western Reserve University, Cleveland, Ohio.,currently affiliated with Fred Hutchinson Cancer Research Center, Division of Oncology, University of Washington School of Medicine, Seattle Cancer Care Alliance, Seattle
| | - Sara M Tolaney
- Department of Medical Oncology, Center of Breast Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lee Schwartzberg
- Division of Hematology/Oncology, The West Clinic, Memphis, Tennessee
| | - Monica Mita
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Georgia McCann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio
| | - Antoinette R Tan
- Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | | | - Andres Forero
- Department of Hematology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carey Anders
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill.,Department of Medicine, University of North Carolina at Chapel Hill
| | - Gerburg M Wulf
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Patrick Dillon
- Division of Hematology/Oncology, University of Virginia, Charlottesville
| | - Filipa Lynce
- Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, DC
| | - Corrine Zarwan
- Department of Hematology and Oncology, Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - John K Erban
- Department of Medicine-Hematology/Oncology, Tufts Medical Center, Boston, Massachusetts
| | | | | | | | | | | | - Melinda L Telli
- Department of Medical Oncology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
555
|
Mehanna J, Haddad FG, Eid R, Lambertini M, Kourie HR. Triple-negative breast cancer: current perspective on the evolving therapeutic landscape. Int J Womens Health 2019; 11:431-437. [PMID: 31447592 PMCID: PMC6682754 DOI: 10.2147/ijwh.s178349] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is known to have a poor prognosis and limited treatment options, namely chemotherapy. Different molecular studies have recently classified TNBC into different subtypes opening the door to potential new-targeted treatment options. In this review, we discuss the current standard of care in the treatment of TNBC in the neoadjuvant, adjuvant and metastatic settings. In addition, we summarize the ongoing phase III clinical trials evaluating different associations between the 3 pillars of anticancer treatment: chemotherapy, targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Joe Mehanna
- Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Fady Gh Haddad
- Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Roland Eid
- Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica Di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Hampig Raphael Kourie
- Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
556
|
Planes-Laine G, Rochigneux P, Bertucci F, Chrétien AS, Viens P, Sabatier R, Gonçalves A. PD-1/PD-L1 Targeting in Breast Cancer: The First Clinical Evidences Are Emerging. A Literature Review. Cancers (Basel) 2019; 11:E1033. [PMID: 31336685 PMCID: PMC6679223 DOI: 10.3390/cancers11071033] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Recently, the development of immunotherapy through the immune checkpoint blockade led to long-lasting responses in several types of cancers that are refractory to conventional treatments, such as melanoma or non-small cell lung cancer. Immunotherapy has also demonstrated significant improvements in various other types of cancers. However, breast cancer remains one of the tumors that have not experienced the explosion of immunotherapy yet. Indeed, breast cancer was traditionally considered as being weakly immunogenic with a lower mutational load compared to other tumor types. In the last few years, anti-PD1/PD-L1 (Programmed death-ligand 1) agents have been evaluated in breast cancer, particularly in the triple negative subtype, with promising results observed when delivered as monotherapy or in combination with conventional treatments. In this review, we will report the results of the most recent studies evaluating immune checkpoint inhibitors in breast cancer. In addition, we will discuss the concomitant development of possible biomarkers, which is required for improving the selection of patients with the highest probability of benefiting from these agents.
Collapse
Affiliation(s)
- Gabrielle Planes-Laine
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Philippe Rochigneux
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
- CRCM-Tumor Immunology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - François Bertucci
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
- CRCM-Predictive Oncology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Anne-Sophie Chrétien
- CRCM-Tumor Immunology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Renaud Sabatier
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
- CRCM-Predictive Oncology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France.
- CRCM-Predictive Oncology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France.
| |
Collapse
|
557
|
Lazarus G, Audrey J, Iskandar AWB. Efficacy and safety profiles of programmed cell death-1/programmed cell death ligand-1 inhibitors in the treatment of triple-negative breast cancer: A comprehensive systematic review. Oncol Rev 2019; 13:425. [PMID: 31857857 PMCID: PMC6886008 DOI: 10.4081/oncol.2019.425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with worse prognosis, with limited treatment regiments available and higher mortality rate. Immune checkpoint inhibitors targeting programmed cell death-1 (PD-1) or programmed cell death-ligand 1 (PD-L1) showed great potentials in treating malignancies and may serve as potential therapies for TNBC. This systematic review aims to evaluate the efficacy and safety profiles of PD-1/PD-L1 inhibitors in the treatment of TNBC. Literature search was performed via PubMed, EBSCOhost, Scopus, and CENTRAL databases, selecting studies which evaluated the use of anti-PD-1/PDL1 for TNBC from inception until February 2019. Risk of bias was assessed by the Newcastle-Ottawa Scale (NOS). Overall, 7 studies evaluating outcomes of 1395 patients with TNBC were included in this systematic review. Anti-PD-1/PD-L1 showed significant antitumor effect, proven by their promising response (objective response rate (ORR), 18.5-39.4%) and survival rates (median overall survival (OS), 9.2-21.3 months). Moreover, anti- PD-1/PD-L1 yielded better outcomes when given as first-line therapy, and overexpression of PD-L1 in tumors showed better therapeutic effects. On the other hands, safety profiles were similar across agents and generally acceptable, with grade ≥3 treatment- related adverse effects (AEs) ranging from 9.5% to 15.6% and no new AEs were experienced by TNBC patients. Most grade ≥3 AEs are immune-mediated, which are manifested as neutropenia, fatigue, peripheral neuropathy, and anemia. PD-1/PD-L1 inhibitors showed promising efficacy and tolerable AEs, and thus may benefit TNBC patients. Further studies of randomized controlled trials with larger populations are needed to better confirm the potential of these agents.
Collapse
|
558
|
Lin FM, Yost SE, Wen W, Frankel PH, Schmolze D, Chu PG, Yuan YC, Liu Z, Yim J, Chen Z, Yuan Y. Differential gene expression and AKT targeting in triple negative breast cancer. Oncotarget 2019; 10:4356-4368. [PMID: 31320990 PMCID: PMC6633890 DOI: 10.18632/oncotarget.27026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/30/2019] [Indexed: 02/03/2023] Open
Abstract
Background: Metastatic triple negative breast cancer (mTNBC) is a heterogeneous disease with poor prognosis. Molecular evolution of TNBC through chemotherapy selection pressure is well recognized but poorly understood. PI3K/AKT/mTOR is one of the most commonly identified oncogenic-driver pathways in breast cancer. The current study is designed to understand the genomic and transcriptomic changes, focusing on the PI3K/AKT/mTOR pathway alterations in paired primary and metastatic TNBCs. Results: Genomic analysis of 7 paired specimens identified 67 known mutations including those from the following signaling pathways: cell cycle, p53, PI3K/AKT/mTOR, RAS/MAPK, and RTK/GF. Principle coordinate analysis (PCoA) identified 4 distinctive molecular groups based on the gene expression patterns of PI3K/AKT/mTOR pathway. Key differentially-expressed genes included AKT3, GSK3B, GNA11, PI3KR1, and GNAQ. Importantly, AKT-targeted therapy showed efficacy in a patient-derived xenograft (PDX) model of TNBC in vivo. Conclusion: Genomic discordance of paired primary and metastatic TNBCs was identified, with significant increase in tumor proliferation pathways seen in metastases. Among the differentially expressed genes, AKT3 can potentially serve as a target for novel combination therapy for treatment of metastatic TNBC. Methods: Paired specimens from 10 patients with TNBCs were identified through an IRB-approved protocol (2002-2015). FoundationOneTM sequencing was performed for genomic profiling, and Affymetrix Human Genechip 2.0st was used for mRNA expression profiling. The similarity among samples was calculated based on Pearson correlation coefficients, which were used to construct hierarchical clustering and heat maps.
Collapse
Affiliation(s)
- Feng-Mao Lin
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Susan E. Yost
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Wei Wen
- Department of Surgery, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Paul H. Frankel
- Department of Biostatistics, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Pei-Guo Chu
- Department of Pathology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Yate-Ching Yuan
- Bioinformatics Core Facility, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Zheng Liu
- Bioinformatics Core Facility, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - John Yim
- Department of Surgery, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Zhen Chen
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| |
Collapse
|
559
|
D'Abreo N, Adams S. Immune-checkpoint inhibition for metastatic triple-negative breast cancer: safety first? Nat Rev Clin Oncol 2019; 16:399-400. [PMID: 31053774 DOI: 10.1038/s41571-019-0216-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Nina D'Abreo
- Oncology Hematology Division, NYU-Winthrop Hospital, Mineola, NY, USA
| | - Sylvia Adams
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, USA.
| |
Collapse
|
560
|
Couey MA, Bell RB, Patel AA, Romba MC, Crittenden MR, Curti BD, Urba WJ, Leidner RS. Delayed immune-related events (DIRE) after discontinuation of immunotherapy: diagnostic hazard of autoimmunity at a distance. J Immunother Cancer 2019; 7:165. [PMID: 31269983 PMCID: PMC6609357 DOI: 10.1186/s40425-019-0645-6] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The risk of delayed autoimmunity occurring months or years after discontinuation of immunotherapy is frequently asserted in the literature. However, specific cases were rarely described until 2018, when a wave of reports surfaced. With expanding I-O indications in the adjuvant/neoadjuvant curative setting, growing numbers of patients will receive limited courses of immunotherapy before entering routine surveillance. In this context, under-recognition of DIRE could pose a growing clinical hazard. METHODS The aim of this study was to characterize DIRE through identification of existing reports of delayed post-treatment irAE in cancer patients treated with immunotherapy. We performed a PubMed literature review from 2008 through 2018 to determine the median data safety reporting window from existing I-O clinical trials, which we then applied to define the DIRE cutoff, and collated all qualifying reports over the same time span. DIRE was defined as new immune-related adverse events (irAE) manifesting ≥90 days after discontinuation of immunotherapy. RESULTS Median duration of I-O clinical trials data safety reporting was 90 days (82% ≤ 90 days). DIRE cutoff was thus set as ≥90 days post-immunotherapy. We identified 23 qualifying cases; 21 by literature review and 2 from our institution. Median off-treatment interval to DIRE was 6 months (range: 3 to 28). Median cumulative immunotherapy exposure was 4 doses (range: 3 to 42). Involvement included endocrine, neurologic, GI, pulmonary, cardiac, rheumatologic and dermatologic irAE. CONCLUSIONS As immunotherapy indications expand into the curative setting, often with brief exposure and potentially sequenced with multimodality treatments, it will be necessary to recognize an emerging diagnostic complex, which we have termed delayed immune-related events (DIRE). Clinical vigilance has the potential to reduce morbidity from diagnostic delay, as irAE are generally manageable with prompt initiation of treatment - or from misdiagnosis - as misattribution can lead to unnecessary or harmful interventions as we describe. DIRE should therefore figure prominently in the differential diagnosis of patients presenting with illnesses of unclear etiology, irrespective of intervening treatments or interval post-immunotherapy, both of which can confound diagnosis. Increased recognition will rest on delineation of DIRE as a clinical diagnostic entity.
Collapse
Affiliation(s)
- Marcus A. Couey
- Robert W. Franz Cancer Center, Providence Portland Medical Center, 2N35 North Pavilion, 4805 N.E. Glisan St, Portland, OR 97213 USA
| | - R. Bryan Bell
- Robert W. Franz Cancer Center, Providence Portland Medical Center, 2N35 North Pavilion, 4805 N.E. Glisan St, Portland, OR 97213 USA
| | - Ashish A. Patel
- Robert W. Franz Cancer Center, Providence Portland Medical Center, 2N35 North Pavilion, 4805 N.E. Glisan St, Portland, OR 97213 USA
| | - Meghan C. Romba
- Providence Neurological Specialties-West, Providence St. Vincent Medical Center, 9135 Southwest Barnes Road, Suite 461, Portland, OR 97225 USA
| | - Marka R. Crittenden
- The Oregon Clinic, Radiation Oncology, 4805 NE Glisan St. Garden Level, Portland, OR 97213 USA
| | - Brendan D. Curti
- Robert W. Franz Cancer Center, Providence Portland Medical Center, 2N35 North Pavilion, 4805 N.E. Glisan St, Portland, OR 97213 USA
| | - Walter J. Urba
- Robert W. Franz Cancer Center, Providence Portland Medical Center, 2N35 North Pavilion, 4805 N.E. Glisan St, Portland, OR 97213 USA
| | - Rom S. Leidner
- Robert W. Franz Cancer Center, Providence Portland Medical Center, 2N35 North Pavilion, 4805 N.E. Glisan St, Portland, OR 97213 USA
| |
Collapse
|
561
|
Wang Y, Zhou S, Yang F, Qi X, Wang X, Guan X, Shen C, Duma N, Vera Aguilera J, Chintakuntlawar A, Price KA, Molina JR, Pagliaro LC, Halfdanarson TR, Grothey A, Markovic SN, Nowakowski GS, Ansell SM, Wang ML. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol 2019; 5:1008-1019. [PMID: 31021376 PMCID: PMC6487913 DOI: 10.1001/jamaoncol.2019.0393] [Citation(s) in RCA: 581] [Impact Index Per Article: 96.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE Programmed cell death (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors have been increasingly used in cancer therapy. Understanding the treatment-related adverse events of these drugs is critical for clinical practice. OBJECTIVE To evaluate the incidences of treatment-related adverse events of PD-1 and PD-L1 inhibitors and the differences between different drugs and cancer types. DATA SOURCES PubMed, Web of Science, Embase, and Scopus were searched from October 1, 2017, through December 15, 2018. STUDY SELECTION Published clinical trials on single-agent PD-1 and PD-L1 inhibitors with tabulated data on treatment-related adverse events were included. DATA EXTRACTION AND SYNTHESIS Trial name, phase, cancer type, PD-1 and PD-L1 inhibitor used, dose escalation, dosing schedule, number of patients, number of all adverse events, and criteria for adverse event reporting data were extracted from each included study, and bayesian multilevel regression models were applied for data analysis. MAIN OUTCOMES AND MEASURES Incidences of treatment-related adverse events and differences between different drugs and cancer types. RESULTS This systematic review and meta-analysis included 125 clinical trials involving 20 128 patients; 12 277 (66.0%) of 18 610 patients from 106 studies developed at least 1 adverse event of any grade (severity), and 2627 (14.0%) of 18 715 patients from 110 studies developed at least 1 adverse event of grade 3 or higher severity. The most common all-grade adverse events were fatigue (18.26%; 95% CI, 16.49%-20.11%), pruritus (10.61%; 95% CI, 9.46%-11.83%), and diarrhea (9.47%; 95% CI, 8.43%-10.58%). The most common grade 3 or higher adverse events were fatigue (0.89%; 95% CI, 0.69%-1.14%), anemia (0.78%; 95% CI, 0.59%-1.02%), and aspartate aminotransferase increase (0.75%; 95% CI, 0.56%-0.99%). Hypothyroidism (6.07%; 95% CI, 5.35%-6.85%) and hyperthyroidism (2.82%; 95% CI, 2.40%-3.29%) were the most frequent all-grade endocrine immune-related adverse events. Nivolumab was associated with higher mean incidences of all-grade adverse events compared with pembrolizumab (odds ratio [OR], 1.28; 95% CI, 0.97-1.79) and grade 3 or higher adverse events (OR, 1.30; 95% CI, 0.89-2.00). PD-1 inhibitors were associated with a higher mean incidence of grade 3 or higher adverse events compared with PD-L1 inhibitors (OR, 1.58; 95% CI, 1.00-2.54). CONCLUSIONS AND RELEVANCE Different PD-1 and PD-L1 inhibitors appear to have varying treatment-related adverse events; a comprehensive summary of the incidences of treatment-related adverse events in clinical trials provides an important guide for clinicians.
Collapse
Affiliation(s)
- Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
- Department of Public Health Sciences, Pennsylvania State College of Medicine, Hershey
| | - Fang Yang
- Medical School of Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xinyue Qi
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Xin Wang
- Medical School of Nanjing University, Nanjing, China
| | - Xiaoxiang Guan
- Medical School of Nanjing University, Nanjing, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chan Shen
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Narjust Duma
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jesus Vera Aguilera
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | - Axel Grothey
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
- West Cancer Center, The University of Tennessee, Memphis
| | | | | | | | - Michael L. Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
562
|
Abstract
Triple-negative breast cancer (TNBC) is characterised by poor outcomes and a historical lack of targeted therapies. Dysregulation of signalling through the phosphoinositide 3 (PI3)-kinase and AKT signalling pathway is one of the most frequent oncogenic aberrations of TNBC. Although mutations in individual genes occur relatively rarely, combined activating mutations in PIK3CA and AKT1, with inactivating mutations in phosphatase and tensin homologue, occur in ∼25%‒30% of advanced TNBC. Recent randomised trials suggest improved progression-free survival (PFS) with AKT-inhibitors in combination with first-line chemotherapy for patients with TNBC and pathway genetic aberrations. We review the evidence for PI3K pathway activation in TNBC, and clinical trial data for PI3K, AKT and mammalian target of rapamycin inhibitors in TNBC. We discuss uncertainty over defining which cancers have pathway activation and the future overlap between immunotherapy and pathway targeting.
Collapse
Affiliation(s)
- J Pascual
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London
| | - N C Turner
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London; Breast Unit, The Royal Marsden Hospital, London, UK.
| |
Collapse
|
563
|
Matikas A, Zerdes I, Lövrot J, Richard F, Sotiriou C, Bergh J, Valachis A, Foukakis T. Prognostic Implications of PD-L1 Expression in Breast Cancer: Systematic Review and Meta-analysis of Immunohistochemistry and Pooled Analysis of Transcriptomic Data. Clin Cancer Res 2019; 25:5717-5726. [PMID: 31227501 DOI: 10.1158/1078-0432.ccr-19-1131] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/15/2019] [Accepted: 06/19/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Conflicting data have been reported on the prognostic value of PD-L1 protein and gene expression in breast cancer.Experimental Design: Medline, Embase, Cochrane Library, and Web of Science Core Collection were searched, and data were extracted independently by two researchers. Outcomes included pooled PD-L1 protein positivity in tumor cells, immune cells, or both, per subtype and per antibody used, and its prognostic value for disease-free and overall survival. A pooled gene expression analysis of 39 publicly available transcriptomic datasets was also performed. RESULTS Of the initial 4,184 entries, 38 retrospective studies fulfilled the predefined inclusion criteria. The overall pooled PD-L1 protein positivity rate was 24% (95% CI, 15%-33%) in tumor cells and 33% (95% CI, 14%- 56%) in immune cells. PD-L1 protein expression in tumor cells was prognostic for shorter overall survival (HR, 1.63; 95% CI, 1.07-2.46; P = 0.02); there was significant heterogeneity (I2 = 80%, P heterogeneity < 0.001). In addition, higher PD-L1 gene expression predicted better survival in multivariate analysis in the entire population (HR, 0.82; 95% CI, 0.74-0.90; P < 0.001 for OS) and in basal-like tumors (HR, 0.64; 95% CI, 0.52-0.80; P < 0.001 for OS; P interaction 0.005). CONCLUSIONS The largest to our knowledge meta-analysis on the subject informs on PD-L1 protein positivity rates and its prognostic value in breast cancer. Standardization is needed prior to routine implementation. PD-L1 gene expression is a promising prognostic factor, especially in basal-like breast cancer. Discrepant prognostic information might be related to PD-L1 gene expression in the stroma.
Collapse
Affiliation(s)
- Alexios Matikas
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden. .,Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Zerdes
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - John Lövrot
- Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Jonas Bergh
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Theodoros Foukakis
- Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
564
|
DNA Repair Deficiency in Breast Cancer: Opportunities for Immunotherapy. JOURNAL OF ONCOLOGY 2019; 2019:4325105. [PMID: 31320901 PMCID: PMC6607732 DOI: 10.1155/2019/4325105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/04/2019] [Accepted: 05/29/2019] [Indexed: 12/19/2022]
Abstract
Historically the development of anticancer treatments has been focused on their effect on tumor cells alone. However, newer treatments have shifted attention to targets on immune cells, resulting in dramatic responses. The effect of DNA repair deficiency on the microenvironment remains an area of key interest. Moreover, established therapies such as DNA damaging treatments such as chemotherapy and PARP inhibitors further modify the tumor microenvironment. Here we describe DNA repair pathways in breast cancer and activation of innate immune pathways in DNA repair deficiency, in particular, the STING (STimulator of INterferon Genes) pathway. Breast tumors with DNA repair deficiency are associated with upregulation of immune checkpoints including PD-L1 (Programmed Death Ligand-1) and may represent a target population for single agent or combination immunotherapy treatment.
Collapse
|
565
|
Liu JF, Gordon M, Veneris J, Braiteh F, Balmanoukian A, Eder JP, Oaknin A, Hamilton E, Wang Y, Sarkar I, Molinero L, Fassò M, O'Hear C, Lin YG, Emens LA. Safety, clinical activity and biomarker assessments of atezolizumab from a Phase I study in advanced/recurrent ovarian and uterine cancers. Gynecol Oncol 2019; 154:314-322. [PMID: 31204078 DOI: 10.1016/j.ygyno.2019.05.021] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/21/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Patients with advanced/recurrent epithelial ovarian and uterine cancers have limited treatment options beyond platinum chemotherapy. Both tumor types can express programmed death-ligand 1 (PD-L1), providing a potential therapeutic target for these patients. Here we present data from the ovarian and uterine cancer cohorts of the Phase I atezolizumab monotherapy study (PCD4989g). METHODS This Phase I, multi-center, first-in-human, open-label, dose-escalation/expansion clinical trial investigated single-agent atezolizumab in cohorts of patients with recurrent epithelial ovarian or uterine cancer. The primary objective was to evaluate the safety and tolerability of single-agent atezolizumab. Anti-tumor activity and preliminary assessment of potential biomarkers were evaluated as secondary and exploratory objectives, respectively. RESULTS The ovarian and uterine cancer cohorts enrolled 12 and 15 patients, respectively (10 [83%] and 5 [33%], respectively, had PD-L1 ≥ 5% on tumor-infiltrating immune cells). Atezolizumab was generally well tolerated with no new safety signals identified. The safety profiles in both cohorts were consistent with the known profile of atezolizumab monotherapy. Treatment-related adverse events (AEs) were mostly Grade ≤ 2, with no treatment-related Grade ≥ 4 AEs reported. Preliminary anti-tumor activity, with long durations of response, was observed in 2 patients from each cohort (ovarian cancer, 8.1 and 30.6+ months; uterine cancer, 7.3 and 16.6+ months). High microsatellite instability and tumor mutational burden were noted in the responders from the uterine cancer cohort. CONCLUSIONS Atezolizumab monotherapy was well tolerated in patients with epithelial ovarian or uterine cancer and may have clinical activity warranting further investigation. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01375842.
Collapse
Affiliation(s)
- Joyce F Liu
- Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215-5450, United States.
| | - Michael Gordon
- HonorHealth Research Institute, 10510 N 92nd St, Suite 200, Scottsdale, AZ 85258, United States.
| | - Jennifer Veneris
- University of Chicago Medicine, 5841 S Maryland Ave, Chicago, IL 60637, United States.
| | - Fadi Braiteh
- Comprehensive Cancer Centers of Nevada, 3730 S Eastern Avenue, Las Vegas, NV 89169, United States.
| | - Ani Balmanoukian
- The Angeles Clinic and Research Institute, 11818 Wilshire Blvd #200, Los Angeles, CA 90025, United States.
| | - Joseph Paul Eder
- Yale Cancer Center, Medical Oncology, PO Box 208028, New Haven, CT 06520-8028, United States.
| | - Ana Oaknin
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Centro Cellex, Calle Natzaret, 115-117, 08035 Barcelona, Spain.
| | - Erika Hamilton
- Tennessee Oncology/Sarah Cannon Research Institute, 250 25th Ave N, Nashville, TN 37203, United States.
| | - Yulei Wang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Indrani Sarkar
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Luciana Molinero
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Marcella Fassò
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Carol O'Hear
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Yvonne G Lin
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Leisha A Emens
- UPMC Hillman Cancer Center, 300 Halket St, Suite 4628, Pittsburgh, PA 15213, United States.
| |
Collapse
|
566
|
Mohan N, Hosain S, Zhao J, Shen Y, Luo X, Jiang J, Endo Y, Wu WJ. Atezolizumab potentiates Tcell-mediated cytotoxicity and coordinates with FAK to suppress cell invasion and motility in PD-L1 + triple negative breast cancer cells. Oncoimmunology 2019; 8:e1624128. [PMID: 31428520 PMCID: PMC6685513 DOI: 10.1080/2162402x.2019.1624128] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/31/2022] Open
Abstract
Immune check point inhibitors targeting programmed cell death protein-1 (PD-1) and its ligand (PD-L1) have shown clinical success in treatment of human malignancies. Triple negative breast cancer (TNBC), which is primarily characterized by high heterogeneity and presence of tumor infiltrating lymphocytes, remains therapeutic challenge due to unavailability of approved targeted therapy. Therapeutic potential of immune check point inhibitors for TNBC patients is under active clinical investigation. In this study, we show that FDA-approved anti-PD-L1 antibody, atezolizumab (ATE), potentiates T cell-mediated cytotoxicity and apoptosis of TNBC cells that express higher levels of PD-L1, but does not have significant effect on TNBC cells expressing low levels of PD-L1. PD-L1 knockdown further confirmed that ability of ATE to promote T cell-induced cytotoxicity is PD-L1 expression dependent. Combination of ATE with PD-L1 upregulating agents, such as HDAC, proteasomal, and lysosomal inhibitors, further augmented cytotoxic activity of T cells toward TNBC cells. Based on analysis of breast cancer tissue samples deposited in The Cancer Genome Atlas (TCGA), we found a positive correlation between PD-L1 and focal adhesion kinase (FAK) mRNA expression in PD-L1-positive (PD-L1+) TNBC, suggesting a functional association of FAK and immune checkpoints. We further demonstrate that ATE dramatically downregulates phosphorylation status of FAK, an important regulator of cell invasion and migration, and significantly enhances FAK inhibitor mediated inhibition of cell motility and invasion of PD-L1+ TNBC cells independent of T cells. Taken together, our data suggest that ATE shows promising anti-tumor activity in PD-L1+ TNBC via both T cell-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Nishant Mohan
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Salman Hosain
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jun Zhao
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA.,Interagency Oncology Task Force (IOTF) Fellowship: Oncology Product Research/Review Fellow, National Cancer Institute, Bethesda, Maryland, USA
| | - Yi Shen
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xiao Luo
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiangsong Jiang
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yukinori Endo
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Wen Jin Wu
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
567
|
Relevance of tumour-infiltrating lymphocytes, PD-1 and PD-L1 in patients with high-risk, nodal-metastasised breast cancer of the German Adjuvant Intergroup Node–positive study. Eur J Cancer 2019; 114:76-88. [DOI: 10.1016/j.ejca.2019.04.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/02/2019] [Accepted: 04/07/2019] [Indexed: 01/07/2023]
|
568
|
Immune induction strategies in metastatic triple-negative breast cancer to enhance the sensitivity to PD-1 blockade: the TONIC trial. Nat Med 2019; 25:920-928. [PMID: 31086347 DOI: 10.1038/s41591-019-0432-4] [Citation(s) in RCA: 624] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/19/2019] [Indexed: 12/19/2022]
Abstract
The efficacy of programmed cell death protein 1 (PD-1) blockade in metastatic triple-negative breast cancer (TNBC) is low1-5, highlighting a need for strategies that render the tumor microenvironment more sensitive to PD-1 blockade. Preclinical research has suggested immunomodulatory properties for chemotherapy and irradiation6-13. In the first stage of this adaptive, non-comparative phase 2 trial, 67 patients with metastatic TNBC were randomized to nivolumab (1) without induction or with 2-week low-dose induction, or with (2) irradiation (3 × 8 Gy), (3) cyclophosphamide, (4) cisplatin or (5) doxorubicin, all followed by nivolumab. In the overall cohort, the objective response rate (ORR; iRECIST14) was 20%. The majority of responses were observed in the cisplatin (ORR 23%) and doxorubicin (ORR 35%) cohorts. After doxorubicin and cisplatin induction, we detected an upregulation of immune-related genes involved in PD-1-PD-L1 (programmed death ligand 1) and T cell cytotoxicity pathways. This was further supported by enrichment among upregulated genes related to inflammation, JAK-STAT and TNF-α signaling after doxorubicin. Together, the clinical and translational data of this study indicate that short-term doxorubicin and cisplatin may induce a more favorable tumor microenvironment and increase the likelihood of response to PD-1 blockade in TNBC. These data warrant confirmation in TNBC and exploration of induction treatments prior to PD-1 blockade in other cancer types.
Collapse
|
569
|
Marra A, Viale G, Curigliano G. Recent advances in triple negative breast cancer: the immunotherapy era. BMC Med 2019; 17:90. [PMID: 31068190 PMCID: PMC6507064 DOI: 10.1186/s12916-019-1326-5] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Several accomplishments have been achieved in triple-negative breast cancer (TNBC) research over the last year. The phase III IMpassion130 trial comparing chemotherapy plus atezolizumab versus chemotherapy plus placebo brought breast cancer into the immunotherapy era. Nevertheless, despite encouraging results being obtained in this trial, many open questions remain. MAIN BODY A positive overall survival outcome was achieved only in PD-L1+ TNBC patients, suggesting a need to enrich the patient population more likely to benefit from an immunotherapeutic approach. Moreover, it remains unknown whether single-agent immunotherapy might be a good option for some patients. In this context, the discovery and implementation of novel and appropriate biomarkers are required. Focusing on the early onset of TNBC, neoadjuvant trials could represent excellent in vivo platforms to test immunotherapy agents and their potential combinations, allowing the performance of translational studies for biomarker implementation and improved patient selection. CONCLUSION The aim of our review is to present recent advances in TNBC treatment and to discuss open issues in order to better define potential future directions for immunotherapy in TNBC.
Collapse
Affiliation(s)
- Antonio Marra
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology (IEO), IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milano, Milano, Italy
| | - Giulia Viale
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology (IEO), IRCCS, Milan, Italy.,Department of Oncology and Haematology, University of Milano, Milano, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology (IEO), IRCCS, Milan, Italy. .,Department of Oncology and Haematology, University of Milano, Milano, Italy.
| |
Collapse
|
570
|
Wang H, Milberg O, Bartelink IH, Vicini P, Wang B, Narwal R, Roskos L, Santa-Maria CA, Popel AS. In silico simulation of a clinical trial with anti-CTLA-4 and anti-PD-L1 immunotherapies in metastatic breast cancer using a systems pharmacology model. ROYAL SOCIETY OPEN SCIENCE 2019; 6:190366. [PMID: 31218069 PMCID: PMC6549962 DOI: 10.1098/rsos.190366] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/24/2019] [Indexed: 05/10/2023]
Abstract
The low response rate of immune checkpoint blockade in breast cancer has highlighted the need for predictive biomarkers to identify responders. While a number of clinical trials are ongoing, testing all possible combinations is not feasible. In this study, a quantitative systems pharmacology model is built to integrate immune-cancer cell interactions in patients with breast cancer, including central, peripheral, tumour-draining lymph node (TDLN) and tumour compartments. The model can describe the immune suppression and evasion in both TDLN and the tumour microenvironment due to checkpoint expression, and mimic the tumour response to checkpoint blockade therapy. We investigate the relationship between the tumour response to checkpoint blockade therapy and composite tumour burden, PD-L1 expression and antigen intensity, including their individual and combined effects on the immune system, using model-based simulations. The proposed model demonstrates the potential to make predictions of tumour response of individual patients given sufficient clinical measurements, and provides a platform that can be further adapted to other types of immunotherapy and their combination with molecular-targeted therapies. The patient predictions demonstrate how this systems pharmacology model can be used to individualize immunotherapy treatments. When appropriately validated, these approaches may contribute to optimization of breast cancer treatment.
Collapse
Affiliation(s)
- Hanwen Wang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Oleg Milberg
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Imke H. Bartelink
- Department of Medicine, University of California, San Francisco, CA, USA
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD), MedImmune, South San Francisco, CA, USA
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Paolo Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK, MedImmune, Cambridge, UK
| | - Bing Wang
- Amador Bioscience Inc, Pleasanton, CA 94588, USA
| | - Rajesh Narwal
- Clinical Pharmacology and DMPK (CPD), MedImmune, Gaithersburg, MD, USA
| | - Lorin Roskos
- Clinical Pharmacology and DMPK (CPD), MedImmune, Gaithersburg, MD, USA
| | - Cesar A. Santa-Maria
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
571
|
Cortés J, André F, Gonçalves A, Kümmel S, Martín M, Schmid P, Schuetz F, Swain SM, Easton V, Pollex E, Deurloo R, Dent R. IMpassion132 Phase III trial: atezolizumab and chemotherapy in early relapsing metastatic triple-negative breast cancer. Future Oncol 2019; 15:1951-1961. [PMID: 30977385 DOI: 10.2217/fon-2019-0059] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The PD-L1 inhibitor atezolizumab received US FDA accelerated approval as treatment for PD-L1-positive metastatic triple-negative breast cancer (TNBC). In IMpassion130, combining atezolizumab with first-line nab-paclitaxel for metastatic TNBC significantly improved progression-free survival and showed a clinically meaningful effect on overall survival in patients with PD-L1-positive tumors. The placebo-controlled randomized Phase III IMpassion132 (NCT03371017) trial is evaluating atezolizumab with first-line chemotherapy (capecitabine [mandatory in platinum-pretreated patients] or gemcitabine/carboplatin) for inoperable locally advanced/metastatic TNBC recurring ≤12 months after completing standard (neo)adjuvant anthracycline and taxane chemotherapy. Stratification factors are: visceral metastases, tumor immune cell PD-L1 status and selected chemotherapy. Patients are randomized to atezolizumab 1200 mg or placebo every 3 weeks with the chosen chemotherapy, continued until progression, unacceptable toxicity or withdrawal. The primary end point is overall survival.
Collapse
Affiliation(s)
- Javier Cortés
- IOB Institute of Oncology, Quironsalud Group, Madrid & Barcelona, & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Fabrice André
- Institut Gustave Roussy, Université Paris Sud, 114, rue Edouard Vaillant, 94800 Villejuif, France
| | - Anthony Gonçalves
- Aix-Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), & Institut Paoli-Calmettes, Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), 232 Bvd Sainte-Marguerite, 13009 Marseille, France
| | - Sherko Kümmel
- Breast Unit Essen, Kliniken Essen-Mitte, Henricistrasse 92, 45136 Essen, Germany
| | - Miguel Martín
- Instituto de Investigación Sanitaria Gregorio Marañón, CiberOnc, Universidad Complutense, Madrid, Spain
| | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, Old Anatomy Building, Charterhouse Square, London EC1M 6BQ, UK
| | - Florian Schuetz
- University Breast Unit, National Center for Tumor Diseases, 69120 Heidelberg, Germany
| | - Sandra M Swain
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 4000 Reservoir Road NW, 120 Building D, Washington, DC 20057, USA
| | - Valerie Easton
- F Hoffmann-La Roche, Hochstrasse, CH 4070 Basel, Switzerland
| | - Erika Pollex
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Regula Deurloo
- F Hoffmann-La Roche, Building 1, Grenzacherstrasse, CH 4070 Basel, Switzerland
| | - Rebecca Dent
- Department of Medical Oncology, National Cancer Center, 11 Hospital Drive, Singapore, 169690, Singapore
| |
Collapse
|
572
|
Yu WD, Sun G, Li J, Xu J, Wang X. Mechanisms and therapeutic potentials of cancer immunotherapy in combination with radiotherapy and/or chemotherapy. Cancer Lett 2019; 452:66-70. [PMID: 30902563 DOI: 10.1016/j.canlet.2019.02.048] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
Immunotherapies based on T cells have gained significant success in the treatment of diverse cancers, however, several limitations also exist, including low response, acquired resistance and severe side effects, which lead to unfavorable outcomes. Recent studies found that traditional therapies, radiotherapy and/or chemotherapy may affect the immune condition in situ and cause abscopal effect, which may improve the response of immunotherapies, enhance the efficiency, and reduce the untoward effect. Here, we review the mechanisms uncovering the cancer immunotherapy and immunogenic effects of radiotherapy and chemotherapy, aiming to highlight the principles underlying the therapeutic potentials of cancer immunotherapy in combination with radiotherapy and/or chemotherapy and ultimately guide better designs for future synergistic cancer therapies.
Collapse
Affiliation(s)
- Wei-Di Yu
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Guan Sun
- Department of Neurosurgery, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu Province, PR China
| | - Jun Li
- Department of General Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, 168 Gushan Road, Nanjing, Jiangsu Province, PR China
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China.
| | - Xiaochen Wang
- Liver Transplantation Center of the First Affiliated Hospital and Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, PR China.
| |
Collapse
|
573
|
Adams S, Diamond JR, Hamilton E, Pohlmann PR, Tolaney SM, Chang CW, Zhang W, Iizuka K, Foster PG, Molinero L, Funke R, Powderly J. Atezolizumab Plus nab-Paclitaxel in the Treatment of Metastatic Triple-Negative Breast Cancer With 2-Year Survival Follow-up: A Phase 1b Clinical Trial. JAMA Oncol 2019; 5:334-342. [PMID: 30347025 PMCID: PMC6439843 DOI: 10.1001/jamaoncol.2018.5152] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/05/2018] [Indexed: 12/30/2022]
Abstract
Importance The humanized monoclonal antibody atezolizumab targets programmed death-ligand 1 and has demonstrated durable single-agent activity in a subset of metastatic triple-negative breast cancers. To extend the observed activity, combinatorial approaches are being tested with standard cytotoxic chemotherapies known to induce immunogenic tumor cell death. Objective To examine the safety, tolerability, and preliminary clinical activity of atezolizumab plus nab-paclitaxel in metastatic triple-negative breast cancers. Design, Setting, and Participants This phase 1b multicohort study enrolled 33 women with stage IV or locally recurrent triple-negative breast cancers and 0 to 2 lines of prior chemotherapy in the metastatic setting from December 8, 2014, to April 30, 2017, at 11 sites in the United States. The median follow-up was 24.4 months (95% CI, 22.1-28.8 months). Interventions Patients received concurrent intravenous atezolizumab and intravenous nab-paclitaxel (minimum 4 cycles). Main Outcomes and Measures The primary end point was safety and tolerability. Secondary end points included best overall response rate by Response Evaluation Criteria in Solid Tumors, version 1.1; objective response rate; duration of response; disease control rate; progression-free survival; overall survival; and biomarker analyses. Results The 33 women had a median age of 55 years (range, 32-84 years) and received 1 or more doses of atezolizumab. All patients (100%) experienced at least 1 treatment-related adverse event, 24 patients (73%) experienced grade 3/4 adverse events, and 7 patients (21%) had grade 3/4 adverse events of special interest. No deaths were related to study treatment. The objective response rate was 39.4% (95% CI, 22.9%-57.9%), and the median duration of response was 9.1 months (95% CI, 2.0-20.9 months). The disease control rate was 51.5% (95% CI, 33.5%-69.2%). Median progression-free survival and overall survival were 5.5 months (95% CI, 5.1-7.7 months) and 14.7 months (95% CI, 10.1-not estimable), respectively. Concurrent nab-paclitaxel neither significantly changed biomarkers of the tumor immune microenvironment (programmed death-ligand 1, tumor-infiltrating lymphocytes, CD8) nor impaired atezolizumab systemic immune activation (expansion of proliferating CD8+ T cells, increase of CXCL10 chemokine). Conclusions and Relevance In this phase 1b trial for metastatic triple-negative breast cancers, the combination of atezolizumab plus nab-paclitaxel had a manageable safety profile. Antitumor responses were observed, including in patients previously treated with a taxane. Trial Registration ClinicalTrials.gov identifier: NCT01633970.
Collapse
Affiliation(s)
- Sylvia Adams
- New York University Perlmutter Cancer Center, New York
| | | | - Erika Hamilton
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville
| | - Paula R. Pohlmann
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | | | | | - Wei Zhang
- Genentech Inc, South San Francisco, California
| | - Koho Iizuka
- Genentech Inc, South San Francisco, California
| | | | | | - Roel Funke
- Genentech Inc, South San Francisco, California
| | - John Powderly
- Carolina BioOncology Institute, Huntersville, North Carolina
| |
Collapse
|
574
|
Pinto JA, Saravia CH, Flores C, Araujo JM, Martínez D, Schwarz LJ, Casas A, Bravo L, Zavaleta J, Chuima B, Alvarado H, Fujita R, Gómez HL. Precision medicine for locally advanced breast cancer: frontiers and challenges in Latin America. Ecancermedicalscience 2019; 13:896. [PMID: 30792813 PMCID: PMC6372295 DOI: 10.3332/ecancer.2019.896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Indexed: 12/18/2022] Open
Abstract
Advances in high-throughput technologies and their involvement in the 'omics' of cancer have made possible the identification of hundreds of biomarkers and the development of predictive and prognostic platforms that model the management of cancer from evidence-based medicine to precision medicine. Latin America (LATAM) is a region characterised by fragmented healthcare, high rates of poverty and disparities to access to a basic standard of care not only for cancer but also for other complex diseases. Patients from the public setting cannot afford targeted therapy, the facilities offering genomic platforms are scarce and the use of high-precision radiotherapy is limited to few facilities. Despite the fact that LATAM oncologists are well-trained in the use of genomic platforms and constantly participate in genomic projects, a medical practice based in precision oncology is a great challenge and frequently limited to private practice. In breast cancer, we are waiting for the results of large basket trials to incorporate the detection of actionable mutations to select targeted treatments, in a similar way to the management of lung cancer. On the other hand and paradoxically, in the 'one fit is not for all' era, clinical and genomic studies continue grouping our patients under the single label 'Latin American' or 'Hispanic' despite the different ancestries and genomic backgrounds seen in the region. More regional cancer genomic initiatives and public availability of this data are needed in order to develop more precise oncology in locally advanced breast cancer.
Collapse
Affiliation(s)
- Joseph A Pinto
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - César H Saravia
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - Claudio Flores
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - Jhajaira M Araujo
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15036, Perú
| | - David Martínez
- Departamento de Radioterapia, Oncosalud-AUNA, Lima 15036, Perú
| | - Luis J Schwarz
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Lima 15036, Perú
| | - Alberto Casas
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima 15067, Perú
| | - Leny Bravo
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima 15067, Perú
| | - Jenny Zavaleta
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima 15067, Perú
| | | | - Hober Alvarado
- Facultad de Ciencias Biológicas, Universidad Nacional San Luis Gonzaga de Ica, Ica 11004, Perú
| | - Ricardo Fujita
- Centro de Genética y Biología Molecular, Universidad de San Martín de Porres, Lima 15024, Perú
| | - Henry L Gómez
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Lima 15036, Perú.,Departamento de Medicina Oncológica, Instituto Nacional de Enfermedades Neoplásicas, 15038, Perú
| |
Collapse
|
575
|
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is associated with poor prognosis and limited treatment options. However, TNBC is known to be more immunogenic compared to other breast cancer subtypes, with tumor-infiltrating lymphocytes playing an important prognostic and predictive role. Furthermore, TNBC has a higher level of programmed cell death-ligand 1 (PD-L1) expression. Therapeutic blockade of PD-L1 using atezolizumab is thus expected to activate and enhance tumor-specific T-cell responses, resulting in improved anti-tumor activity. Areas covered: This review summarizes the development and the impact of the PD-L1 inhibitor atezolizumab in advanced TNBC; it examines the mechanism of action, pharmacokinetics and the available preclinical and clinical data. Expert opinion: Atezolizumab, a novel immune checkpoint inhibitors targeting PD-L1, is an effective and well-tolerated treatment option for metastatic TNBC. In general, TNBC has a high unmet medical need, hence the clinical development of atezolizumab should continue, particularly for TNBC. Indeed, atezolizumab has the potential to substantially augment the therapeutic armamentarium for TNBC. This should lead to improved immunotherapeutic strategies and the enhancement of the outcome for this group of breast cancer patients.
Collapse
Affiliation(s)
- Anne-Sophie Heimes
- a Department of Obstetrics and Gynecology, Division of Molecular Oncology , University Medical Center , Mainz , Germany
| | - Marcus Schmidt
- a Department of Obstetrics and Gynecology, Division of Molecular Oncology , University Medical Center , Mainz , Germany
| |
Collapse
|
576
|
Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Hegg R, Im SA, Shaw Wright G, Henschel V, Molinero L, Chui SY, Funke R, Husain A, Winer EP, Loi S, Emens LA. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N Engl J Med 2018; 379:2108-2121. [PMID: 30345906 DOI: 10.1056/nejmoa1809615] [Citation(s) in RCA: 2964] [Impact Index Per Article: 423.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Unresectable locally advanced or metastatic triple-negative (hormone-receptor-negative and human epidermal growth factor receptor 2 [HER2]-negative) breast cancer is an aggressive disease with poor outcomes. Nanoparticle albumin-bound (nab)-paclitaxel may enhance the anticancer activity of atezolizumab. METHODS In this phase 3 trial, we randomly assigned (in a 1:1 ratio) patients with untreated metastatic triple-negative breast cancer to receive atezolizumab plus nab-paclitaxel or placebo plus nab-paclitaxel; patients continued the intervention until disease progression or an unacceptable level of toxic effects occurred. Stratification factors were the receipt or nonreceipt of neoadjuvant or adjuvant taxane therapy, the presence or absence of liver metastases at baseline, and programmed death ligand 1 (PD-L1) expression at baseline (positive vs. negative). The two primary end points were progression-free survival (in the intention-to-treat population and PD-L1-positive subgroup) and overall survival (tested in the intention-to-treat population; if the finding was significant, then it would be tested in the PD-L1-positive subgroup). RESULTS Each group included 451 patients (median follow-up, 12.9 months). In the intention-to-treat analysis, the median progression-free survival was 7.2 months with atezolizumab plus nab-paclitaxel, as compared with 5.5 months with placebo plus nab-paclitaxel (hazard ratio for progression or death, 0.80; 95% confidence interval [CI], 0.69 to 0.92; P=0.002); among patients with PD-L1-positive tumors, the median progression-free survival was 7.5 months and 5.0 months, respectively (hazard ratio, 0.62; 95% CI, 0.49 to 0.78; P<0.001). In the intention-to-treat analysis, the median overall survival was 21.3 months with atezolizumab plus nab-paclitaxel and 17.6 months with placebo plus nab-paclitaxel (hazard ratio for death, 0.84; 95% CI, 0.69 to 1.02; P=0.08); among patients with PD-L1-positive tumors, the median overall survival was 25.0 months and 15.5 months, respectively (hazard ratio, 0.62; 95% CI, 0.45 to 0.86). No new adverse effects were identified. Adverse events that led to the discontinuation of any agent occurred in 15.9% of the patients who received atezolizumab plus nab-paclitaxel and in 8.2% of those who received placebo plus nab-paclitaxel. CONCLUSIONS Atezolizumab plus nab-paclitaxel prolonged progression-free survival among patients with metastatic triple-negative breast cancer in both the intention-to-treat population and the PD-L1-positive subgroup. Adverse events were consistent with the known safety profiles of each agent. (Funded by F. Hoffmann-La Roche/Genentech; IMpassion130 ClinicalTrials.gov number, NCT02425891 .).
Collapse
Affiliation(s)
- Peter Schmid
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Sylvia Adams
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Hope S Rugo
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Andreas Schneeweiss
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Carlos H Barrios
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Hiroji Iwata
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Véronique Diéras
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Roberto Hegg
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Seock-Ah Im
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Gail Shaw Wright
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Volkmar Henschel
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Luciana Molinero
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Stephen Y Chui
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Roel Funke
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Amreen Husain
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Eric P Winer
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Sherene Loi
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| | - Leisha A Emens
- From the Barts Cancer Institute, Queen Mary University of London, London (P.S.); Perlmutter Cancer Center, New York University School of Medicine, New York (S.A.); the Department of Medicine, University of California, San Francisco, San Francisco (H.S.R.), and Genentech, South San Francisco (L.M., S.Y.C., R.F.) - both in California; University Hospital Heidelberg, Heidelberg, Germany (A.S.); Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre (C.H.B.), and the University of São Paulo, São Paulo (R.H.) - both in Brazil; Aichi Cancer Center Hospital, Nagoya, Japan (H.I.); the Department of Medical Oncology, Institut Curie, Paris (V.D.); Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea (S.-A.I.); Florida Cancer Specialists and Research Institute, New Port Richey (G.S.W.); Roche, Basel, Switzerland (V.H., A.H.); Dana-Farber Cancer Institute, Boston (E.P.W.); Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia (S.L.); and the Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore (L.A.E.)
| |
Collapse
|
577
|
Abstract
PURPOSE OF REVIEW This review summarizes current immunotherapies in breast cancer, with an emphasis on immune checkpoint inhibitors and vaccines. RECENT FINDINGS Combination immunotherapy with checkpoint inhibitors and cytotoxic therapies have shown promising results. Active clinical trials are ongoing in both early stage and metastatic settings for triple negative, HER2+, and hormone-positive breast cancer patients. SUMMARY Ongoing challenges remain in defining biomarkers that predict response to immunotherapy, determining the optimal combination immunotherapies, and enhancing the immunogenicity of breast cancer subtypes.
Collapse
Affiliation(s)
- Zishuo I Hu
- 1Mount Sinai Health System, Icahn School of Medicine, New York, NY USA
| | - Heather L McArthur
- 2Department of Medicine, Breast Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, 1S35, Los Angeles, CA 90048 USA
| |
Collapse
|