501
|
Gonzalez-Ericsson PI, Stovgaard ES, Sua LF, Reisenbichler E, Kos Z, Carter JM, Michiels S, Le Quesne J, Nielsen TO, Laenkholm AV, Fox SB, Adam J, Bartlett JM, Rimm DL, Quinn C, Peeters D, Dieci MV, Vincent-Salomon A, Cree I, Hida AI, Balko JM, Haynes HR, Frahm I, Acosta-Haab G, Balancin M, Bellolio E, Yang W, Kirtani P, Sugie T, Ehinger A, Castaneda CA, Kok M, McArthur H, Siziopikou K, Badve S, Fineberg S, Gown A, Viale G, Schnitt SJ, Pruneri G, Penault-Llorca F, Hewitt S, Thompson EA, Allison KH, Symmans WF, Bellizzi AM, Brogi E, Moore DA, Larsimont D, Dillon DA, Lazar A, Lien H, Goetz MP, Broeckx G, El Bairi K, Harbeck N, Cimino-Mathews A, Sotiriou C, Adams S, Liu SW, Loibl S, Chen IC, Lakhani SR, Juco JW, Denkert C, Blackley EF, Demaria S, Leon-Ferre R, Gluz O, Zardavas D, Emancipator K, Ely S, Loi S, Salgado R, Sanders M. The path to a better biomarker: application of a risk management framework for the implementation of PD-L1 and TILs as immuno-oncology biomarkers in breast cancer clinical trials and daily practice. J Pathol 2020; 250:667-684. [PMID: 32129476 DOI: 10.1002/path.5406] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 02/18/2020] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitor therapies targeting PD-1/PD-L1 are now the standard of care in oncology across several hematologic and solid tumor types, including triple negative breast cancer (TNBC). Patients with metastatic or locally advanced TNBC with PD-L1 expression on immune cells occupying ≥1% of tumor area demonstrated survival benefit with the addition of atezolizumab to nab-paclitaxel. However, concerns regarding variability between immunohistochemical PD-L1 assay performance and inter-reader reproducibility have been raised. High tumor-infiltrating lymphocytes (TILs) have also been associated with response to PD-1/PD-L1 inhibitors in patients with breast cancer (BC). TILs can be easily assessed on hematoxylin and eosin-stained slides and have shown reliable inter-reader reproducibility. As an established prognostic factor in early stage TNBC, TILs are soon anticipated to be reported in daily practice in many pathology laboratories worldwide. Because TILs and PD-L1 are parts of an immunological spectrum in BC, we propose the systematic implementation of combined PD-L1 and TIL analyses as a more comprehensive immuno-oncological biomarker for patient selection for PD-1/PD-L1 inhibition-based therapy in patients with BC. Although practical and regulatory considerations differ by jurisdiction, the pathology community has the responsibility to patients to implement assays that lead to optimal patient selection. We propose herewith a risk-management framework that may help mitigate the risks of suboptimal patient selection for immuno-therapeutic approaches in clinical trials and daily practice based on combined TILs/PD-L1 assessment in BC. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Elisabeth S Stovgaard
- Department of Pathology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Luz F Sua
- Department of Pathology and Laboratory Medicine, Fundación Valle del Lili, and Faculty of Health Sciences, Universidad ICESI, Cali, Colombia
| | | | - Zuzana Kos
- Department of Pathology, BC Cancer Agency, Vancouver, Canada
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Stefan Michiels
- Biostatistics and Epidemiology Service, Centre de Recherche en Epidémiologie et Santé des Populations, Gustave Roussy, Université Paris-Sud, Villejuif, France
| | - John Le Quesne
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- MRC Toxicology Unit, University of Cambridge, Leicester, UK
| | - Torsten O Nielsen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | | | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Julien Adam
- Department of Pathology, Gustave Roussy, Grand Paris, France
| | - John Ms Bartlett
- Ontario Institute for Cancer Research, Toronto, Canada
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Cecily Quinn
- Department of Pathology, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | - Dieter Peeters
- HistoGeneX NV, Antwerp, Belgium
- AZ Sint-Maarten Hospital, Mechelen, Belgium
| | - Maria V Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto - IRCCS, Padova, Italy
| | | | - Ian Cree
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France
| | - Akira I Hida
- Department of Pathology, Matsuyama Shimin Hospital, Matsuyama, Japan
| | - Justin M Balko
- Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Harry R Haynes
- Department of Cellular Pathology, North Bristol NHS Trust, Bristol, UK
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Isabel Frahm
- Department of Pathology, Sanatorio Mater Dei, Buenos Aires, Argentina
| | - Gabriela Acosta-Haab
- Department of Pathology, Hospital de Oncología Maria Curie, Buenos Aires, Argentina
| | - Marcelo Balancin
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Enrique Bellolio
- Department of Pathology, Universidad de La Frontera, Temuco, Chile
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, PR China
| | - Pawan Kirtani
- Department of Histopathology, Manipal Hospitals Dwarka, New Delhi, India
| | - Tomoharu Sugie
- Breast Surgery, Kansai Medical University Hospital, Hirakata, Japan
| | - Anna Ehinger
- Department of Clinical Genetics and Pathology, Skane University Hospital, Lund University, Lund, Sweden
| | - Carlos A Castaneda
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Marleen Kok
- Divisions of Medical Oncology, Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Heather McArthur
- Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kalliopi Siziopikou
- Department of Pathology, Breast Pathology Section, Northwestern University, Chicago, IL, USA
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| | - Susan Fineberg
- Department of Pathology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, USA
| | - Allen Gown
- PhenoPath Laboratories, Seattle, WA, USA
| | - Giuseppe Viale
- Department of Pathology, Istituto Europeo di Oncologia IRCCS, Milan, Italy
- University of Milan, Milan, Italy
| | - Stuart J Schnitt
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Giancarlo Pruneri
- University of Milan, Milan, Italy
- Department of Pathology, IRCCS Fondazione Instituto Nazionale Tumori, Milan, Italy
| | - Frederique Penault-Llorca
- Department of Biology and Pathology, Centre Jean Perrin, Clermont Ferrand, France
- UMR INSERM 1240, Université Clermont Auvergne, Clermont Ferrand, France
| | - Stephen Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - William F Symmans
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew M Bellizzi
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David A Moore
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, and Department of Cellular Pathology, UCLH, London, UK
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Deborah A Dillon
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander Lazar
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Huangchun Lien
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | | | - Glenn Broeckx
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Khalid El Bairi
- Cancer Biomarkers Working Group, Faculty of Medicine and Pharmacy, Mohamed Ist University, Oujda, Morocco
| | - Nadia Harbeck
- Breast Center, Department of OB&GYN and CCC (LMU), University of Munich, Munich, Germany
| | - Ashley Cimino-Mathews
- Department of Pathology and Oncology, The Johns Hopkins Hospital, Baltimore, MD, USA
| | - Christos Sotiriou
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sylvia Adams
- Perlmutter Cancer Center, New York University Medical School, New York, NY, USA
| | | | | | - I-Chun Chen
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Sunil R Lakhani
- The University of Queensland, Centre for Clinical Research, and Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, Australia
| | - Jonathan W Juco
- Translational Medicine, Merck & Co, Inc, Kenilworth, NJ, USA
| | - Carsten Denkert
- Institute of Pathology, Universitätsklinikum Gießen und Marburg GmbH, Standort Marburg and Philipps-Universität Marburg, Marburg, Germany
| | - Elizabeth F Blackley
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Sandra Demaria
- Department of Radiation Oncology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Oleg Gluz
- Johanniter GmbH - Evangelisches Krankenhaus Bethesda Mönchengladbach, West German Study Group, Mönchengladbach, Germany
| | | | | | - Scott Ely
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Sherene Loi
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Roberto Salgado
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
| | - Melinda Sanders
- Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
502
|
Zhao S, Zuo WJ, Shao ZM, Jiang YZ. Molecular subtypes and precision treatment of triple-negative breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:499. [PMID: 32395543 PMCID: PMC7210152 DOI: 10.21037/atm.2020.03.194] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype. Despite the progress made in precision treatment of cancer patients, targeted treatment is still at its early stage in TNBC, and chemotherapy remains the standard treatment. With the advances in next generation sequencing technology, genomic and transcriptomic analyses have provided deeper insight into the inter-tumoral heterogeneity of TNBC. Much effort has been made to classify TNBCs into different molecular subtypes according to genetic aberrations and expression signatures and to uncover novel treatment targets. In this review, we summarized the current knowledge regarding the molecular classification of TNBC and explore the future paradigm for using molecular classification to guide the development of precision treatment and clinical practice.
Collapse
Affiliation(s)
- Shen Zhao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wen-Jia Zuo
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| |
Collapse
|
503
|
Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, Blair SL, Burstein HJ, Dang C, Elias AD, Giordano SH, Goetz MP, Goldstein LJ, Isakoff SJ, Krishnamurthy J, Lyons J, Marcom PK, Matro J, Mayer IA, Moran MS, Mortimer J, O'Regan RM, Patel SA, Pierce LJ, Rugo HS, Sitapati A, Smith KL, Smith ML, Soliman H, Stringer-Reasor EM, Telli ML, Ward JH, Young JS, Burns JL, Kumar R. Breast Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 18:452-478. [DOI: 10.6004/jnccn.2020.0016] [Citation(s) in RCA: 572] [Impact Index Per Article: 114.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Several new systemic therapy options have become available for patients with metastatic breast cancer, which have led to improvements in survival. In addition to patient and clinical factors, the treatment selection primarily depends on the tumor biology (hormone-receptor status and HER2-status). The NCCN Guidelines specific to the workup and treatment of patients with recurrent/stage IV breast cancer are discussed in this article.
Collapse
Affiliation(s)
| | | | - Jame Abraham
- 3Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | - Rebecca Aft
- 4Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Doreen Agnese
- 5The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | | | | | - Chau Dang
- 9Memorial Sloan Kettering Cancer Center
| | | | | | | | | | | | | | - Janice Lyons
- 3Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Jennifer Matro
- 17Abramson Cancer Center at the University of Pennsylvania
| | | | | | | | | | | | | | - Hope S. Rugo
- 23UCSF Helen Diller Family Comprehensive Cancer Center
| | | | - Karen Lisa Smith
- 24The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | - John H. Ward
- 28Huntsman Cancer Institute at the University of Utah
| | | | | | | |
Collapse
|
504
|
Keenan TE, Tolaney SM. Role of Immunotherapy in Triple-Negative Breast Cancer. J Natl Compr Canc Netw 2020; 18:479-489. [DOI: 10.6004/jnccn.2020.7554] [Citation(s) in RCA: 365] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have led to durable clinical remissions in many metastatic cancers. However, the single-agent efficacy of ICIs in breast cancer is low, including in triple-negative breast cancer (TNBC), which has several key characteristics that enhance ICI responses. Strategies to improve anticancer immune responses in TNBC are urgently needed to extend survival for patients with metastatic disease. This review presents ICI monotherapy response rates and discusses combination strategies with chemotherapy, targeted therapies, and novel immunotherapies. It concludes with a summary of immunotherapy biomarkers in TNBC and a call to action for future directions of research critical to advancing the efficacy of immunotherapy for patients with TNBC.
Collapse
Affiliation(s)
- Tanya E. Keenan
- 1Department of Medical Oncology, Dana-Farber Cancer Institute, and
- 2Harvard Medical School, Boston, Massachusetts
| | - Sara M. Tolaney
- 1Department of Medical Oncology, Dana-Farber Cancer Institute, and
- 2Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
505
|
Liu ZB, Zhang L, Bian J, Jian J. Combination Strategies of Checkpoint Immunotherapy in Metastatic Breast Cancer. Onco Targets Ther 2020; 13:2657-2666. [PMID: 32308409 PMCID: PMC7133118 DOI: 10.2147/ott.s240655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Checkpoint immunotherapy is emerging as a new therapeutic approach for metastatic breast cancer. Monotherapy of immunoagents against PD1/PD-L1 or CTLA-4 has shown little efficacy in these patients. Recently, to determine the optimal use of immunotherapy, there has been a rapid expansion in the number of clinical trials developing immunotherapy combinations. These combination therapeutic approaches can enhance various aspects of cancer immunity, such as tumor antigenicity or intratumor T cell infiltration, which provides a theoretical basis for combining them with checkpoint immunotherapy to achieve synergistic effects. Here, we review the available data and ongoing efforts to establish the safety and efficacy of immunoagents in combination with chemotherapy, radiotherapy, HER2-targeted therapy, CDK4/6 inhibitors, PARP inhibitors, and another checkpoint immunoagents.
Collapse
Affiliation(s)
- Zhi Bing Liu
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| | - Luyan Zhang
- Department of Oncology, Binzhou People's Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| | - Jia Bian
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| | - Jinbo Jian
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, Shandong Province 256600, People's Republic of China
| |
Collapse
|
506
|
Dadiani M, Necula D, Kahana-Edwin S, Oren N, Baram T, Marin I, Morzaev-Sulzbach D, Pavlovski A, Balint-Lahat N, Anafi L, Wiemann S, Korner C, Gal-Yam EN, Avivi C, Kaufman B, Barshack I, Ben-Baruch A. TNFR2+ TILs are significantly associated with improved survival in triple-negative breast cancer patients. Cancer Immunol Immunother 2020; 69:1315-1326. [PMID: 32198536 DOI: 10.1007/s00262-020-02549-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022]
Abstract
In view of the relatively limited efficacy of immunotherapies targeting the PD-1-PD-L1 axis in triple-negative breast cancer (TNBC) and of published reports on tumor-promoting roles of TNFR2+ tumor-infiltrating lymphocytes (TNFR2+ TILs), we determined the incidence of TNFR2+ TILs in TNBC patient tumors, their association with disease outcome and relations with PD-1+ TILs. Using a cohort of treatment-naïve TNBC patients with long follow-up (n = 70), we determined the presence of TNFR2+ TILs and PD-1+ TILs by immunohistochemistry. TILs (≥ 1% of cellular mass) and TNFR2+ TILs (≥ 1% of total TILs) were detected in 96% and 74% of tumors, respectively. The presence of TILs at > 5% of tumor cell mass ("Positive TILs"), as well as of positive TNFR2+ TILs (> 5%), was independently associated with good prognosis, and combination of both parameters demonstrated superior outcome relative to their lower levels. PD1+ TILs (> 5/hot spot) were detected in 63% of patients. High levels of PD-1+ TILs (> 20/hot spot) showed an unfavorable disease outcome, and in their presence, the favorable outcome of positive TNFR2+ TILs was ablated. Thus, TNFR2+ TILs are strongly connected to improved prognosis in TNBC; these findings suggest that TNFR2+ TILs have favorable effects in TNBC patients, unlike the tumor-promoting roles attributed to them in other cancer systems. Overall, our observations propose that the TNFR2+ TIL subset should not be targeted in the course of TNBC therapy; rather, its beneficial impacts may become into power when anti-PD-1 regimens-that may potentiate immune activities-are administered to TNBC patients.
Collapse
Affiliation(s)
- Maya Dadiani
- Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Daniela Necula
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Nino Oren
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Tamir Baram
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Irina Marin
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Anya Pavlovski
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | | | - Liat Anafi
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Korner
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Camila Avivi
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Bella Kaufman
- Breast Oncology Institute, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Barshack
- Pathology Institute, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adit Ben-Baruch
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
507
|
Simmons CE, Brezden-Masley C, McCarthy J, McLeod D, Joy AA. Positive progress: current and evolving role of immune checkpoint inhibitors in metastatic triple-negative breast cancer. Ther Adv Med Oncol 2020; 12:1758835920909091. [PMID: 33014143 PMCID: PMC7517981 DOI: 10.1177/1758835920909091] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
Background: Triple-negative breast cancer (TNBC) represents an aggressive breast cancer subtype with historically poor overall outcomes, due primarily to a lack of effective targeted agents. Chemotherapy has been the primary treatment approach, although immune checkpoint inhibitors (ICIs) are currently being investigated to improve patient outcomes. This review examines the clinical implications of current evidence on the use of ICIs for the treatment of metastatic TNBC. Methods: Our systematic search identified two phase III and five phase I/II trials reporting on the efficacy of ICIs used as monotherapy or combined with chemotherapy for the treatment of metastatic TNBC. Results: The phase III IMpassion 130 trial showed a significant improvement in median progression-free survival in the intent-to-treat (net 1.7 months, p = 0.002) and PD-L1-positive populations (net 2.5 months, p < 0.001) for the addition of first-line atezolizumab versus placebo to nab-paclitaxel in metastatic TNBC. Although median overall survival was not significantly improved in patients receiving atezolizumab overall [net 2.3 months, hazard ratio (HR) 0.86, 95% confidence interval (CI) 0.72–1.02, p = 0.078], numerical improvements in the PD-L1-positive population were compelling (net 7.0 months, HR 0.71; 95% CI 0.54–0.93). Toxicity profiles were as expected, and no new safety signals were observed. Pembrolizumab monotherapy did not significantly improve overall survival in similar patients that had received prior treatment in KEYNOTE-119. Conclusions: Atezolizumab plus nab-paclitaxel represents a potential new first-line standard of care for patients with metastatic PD-L1-positive TNBC. Other ICIs used as monotherapy, or combined with chemotherapy for advanced TNBC, as well as their use for earlier stage disease, are areas of ongoing investigation.
Collapse
Affiliation(s)
- Christine E Simmons
- Division of Medical Oncology, BC Cancer Agency-Vancouver, 600 West 10th Avenue, Vancouver, British Columbia, V5Z 4E6, Canada
| | | | - Joy McCarthy
- Dr H. Bliss Murphy Cancer Centre, St. John's, Newfoundland, Canada
| | | | | |
Collapse
|
508
|
van Dongen MGJ, Kok M. Mise en place: toward neoadjuvant chemoimmunotherapy for early triple-negative breast cancer. Ann Oncol 2020; 31:556-557. [PMID: 32171753 DOI: 10.1016/j.annonc.2020.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 02/27/2020] [Indexed: 11/26/2022] Open
Affiliation(s)
- M G J van Dongen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M Kok
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Tumor Biology & Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
509
|
Vafaizadeh V, Barekati Z. Immuno-Oncology Biomarkers for Personalized Immunotherapy in Breast Cancer. Front Cell Dev Biol 2020; 8:162. [PMID: 32258038 PMCID: PMC7089925 DOI: 10.3389/fcell.2020.00162] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/28/2020] [Indexed: 12/15/2022] Open
Abstract
The immune checkpoint blockade therapy has drastically advanced treatment of different types of cancer over the past few years. Female breast cancer is the second leading cause of death in the overall burden of cancers worldwide that is encouraging healthcare professionals to improve cancer care management. The checkpoint blockade therapies combined with novel agents become the recent focus of various clinical trials in breast cancer. However, identification of the patients who are responsive to these therapeutic strategies remained as a major issue for enhancing the efficacy of these treatments. This highlights the unmet need in discovery and development of novel biomarkers to add predictive values for prosperous personalized medicine. In this review we summarize the advances done in the era of biomarker studies and highlight their link in supporting breast cancer immunotherapy.
Collapse
Affiliation(s)
- Vida Vafaizadeh
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Zeinab Barekati
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
510
|
He TF, Yost SE, Frankel PH, Dagis A, Cao Y, Wang R, Rosario A, Tu TY, Solomon S, Schmolze D, Mortimer J, Lee P, Yuan Y. Multi-panel immunofluorescence analysis of tumor infiltrating lymphocytes in triple negative breast cancer: Evolution of tumor immune profiles and patient prognosis. PLoS One 2020; 15:e0229955. [PMID: 32150594 PMCID: PMC7062237 DOI: 10.1371/journal.pone.0229955] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
The evolutionary changes in immune profiles of triple negative breast cancer (TNBC) are not well understood, although it is known that immune checkpoint inhibitors have diminished activity in heavily pre-treated TNBC patients. This study was designed to characterize immune profile changes of longitudinal tumor specimens by studying immune subsets of tumor infiltrating lymphocytes (TILs) in paired primary and metastatic TNBC in a cohort of "poor outcome" (relapsed within 5 years) patients. Immune profiles of TNBCs in a cohort of "good outcome" (no relapse within 5 years) patients were also analyzed. Immune subsets were characterized for CD4, CD8, FOXP3, CD20, CD33, and PD1 using immuno-fluorescence staining in stroma, tumor, and combined stroma and tumor tissue. TIL subsets in "good outcome" versus "poor outcome" patients were also analyzed. Compared with primary, metastatic TNBCs had significantly lower TILs by hematoxylin and eosin (H&E) staining. Stromal TILs (sTILs), but not tumoral TILs (tTILs) had significantly reduced cytotoxic CD8+ T cells (CTLs), PD1+ CTLs, and total PD1+ TILs in metastatic compared with matched primary TNBCs. Higher PD1+ CTLs, PD1+CD4+ helper T cells (PD1+TCONV) and all PD1+ T cells in sTILs, tTILs and total stromal and tumor TILS (s+tTIL) were all associated with better prognosis. In summary, TIL subsets decrease significantly in metastatic TNBCs compared with matched primary. Higher PD1+ TILs are associated with better prognosis in early stage TNBCs. This finding supports the application of immune checkpoint inhibitors early in the treatment of TNBCs.
Collapse
Affiliation(s)
- Ting-Fang He
- Department of Immuno-Oncology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Susan E. Yost
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, California, United States of America
| | - Paul H. Frankel
- Department of Biostatistics, City of Hope National Medical Center, Duarte, California, United States of America
| | - Andrew Dagis
- Department of Biostatistics, City of Hope National Medical Center, Duarte, California, United States of America
| | - Yu Cao
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, California, United States of America
| | - Roger Wang
- Department of Immuno-Oncology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Anthony Rosario
- Department of Immuno-Oncology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Travis Yiwey Tu
- Department of Immuno-Oncology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Shawn Solomon
- Department of Immuno-Oncology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Joanne Mortimer
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, California, United States of America
| | - Peter Lee
- Department of Immuno-Oncology, City of Hope National Medical Center, Duarte, California, United States of America
| | - Yuan Yuan
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, California, United States of America
| |
Collapse
|
511
|
Breast cancer vaccines: Heeding the lessons of the past to guide a path forward. Cancer Treat Rev 2020; 84:101947. [DOI: 10.1016/j.ctrv.2019.101947] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 01/29/2023]
|
512
|
Eiger D, Brandão M, de Azambuja E. Lessons learned at SABCS 2019 and to-dos from immunotherapy in breast cancer. ESMO Open 2020; 5:e000688. [PMID: 32188717 PMCID: PMC7078694 DOI: 10.1136/esmoopen-2020-000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 11/04/2022] Open
Affiliation(s)
- Daniel Eiger
- Academic Promoting Team, Institut Jules Bordet et L'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Mariana Brandão
- Academic Promoting Team, Institut Jules Bordet et L'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Evandro de Azambuja
- Academic Promoting Team, Institut Jules Bordet et L'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| |
Collapse
|
513
|
Kagihara JA, Andress M, Diamond JR. Nab-paclitaxel and atezolizumab for the treatment of PD-L1-positive, metastatic triple-negative breast cancer: review and future directions. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:59-65. [PMID: 32190733 DOI: 10.1080/23808993.2020.1730694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction Breast cancer is the most common malignancy in women in the United States and triple-negative breast cancer (TNBC) accounts for 15-20%. The standard of care for metastatic TNBC has been limited to cytotoxic chemotherapy with modest efficacy. TNBC is associated with high levels of tumor-infiltrating lymphocytes and PD-L1 expression, supporting the investigation of immune checkpoint inhibitors in this breast cancer subtype. Areas Covered This review summarizes the clinical data supporting the use of atezolizumab and nab-paclitaxel in the treatment of metastatic PD-L1-positive TNBC. It examines the pharmacology and toxicity profile of the combination in patients with metastatic TNBC. Expert Opinion The addition of atezolizumab to nab-paclitaxel prolonged progression-free survival in both the intention-to-treat and PD-L1-positive subgroups in the first line setting in patients with metastatic TNBC. The IMpassion 130 trial led to FDA-approval of this combination in patients with PD-L1-positive, metastatic TNBC and represents the first approval of immunotherapy for TNBC. This work supports ongoing investigations of other immunotherapy combinations in TNBC, predictive biomarker development and immunotherapy in patients with early stage TNBC. Immunotherapy combinations in TNBC have the potential to lead to improved survival in this group of patients with high risk disease.
Collapse
Affiliation(s)
- Jodi A Kagihara
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| | - Michelle Andress
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| | - Jennifer R Diamond
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, United States of America
| |
Collapse
|
514
|
Rossi G, Zullo L, Cerbone L, Coco S, Longo L, Tagliamento M, Dal Bello MG, Boccardo S, Alama A, Genova C. ADP ribose polymerase inhibitors for treating non-small cell lung cancer: new additions to the pharmacotherapeutic armamentarium. Expert Opin Pharmacother 2020; 21:679-686. [PMID: 32073315 DOI: 10.1080/14656566.2020.1724283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Poly (ADP-ribose) polymerase inhibitors (PARPi) are already part of the armamentarium of drugs available against ovarian and breast cancer. There is less data available on the efficacy of these drugs in the treatment of non-small cell lung cancer (NSCLC). AREAS COVERED The authors have analyzed the preclinical studies that justified the use of PARPi in NSCLC. They then evaluate the in vivo efficacy of the combination of these drugs with chemotherapy, radiotherapy, and immunotherapy. EXPERT OPINION Data from clinical trials available to date have discouraged the use of PARPi in association with chemotherapy or radiotherapy in NSCLC. The knowledge available to date opens the door to the use of PARPi in association with immunotherapy. In fact, the activity of these drugs would not be based only on direct cytotoxic action, but also on the modification of the intra-tumor microenvironment, in particular by increasing the expression of PD-L1 on tumor cells. This action might potentially enhance available treatments with a modest increase in toxicity.
Collapse
Affiliation(s)
- Giovanni Rossi
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Medical, Surgical and Experimental Sciences, University of Sassari , Italy
| | - Lodovica Zullo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Luigi Cerbone
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Luca Longo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Marco Tagliamento
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | | | - Simona Boccardo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Angela Alama
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Carlo Genova
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| |
Collapse
|
515
|
Ho AY, Barker CA, Arnold BB, Powell SN, Hu ZI, Gucalp A, Lebron-Zapata L, Wen HY, Kallman C, D'Agnolo A, Zhang Z, Flynn J, Dunn SA, McArthur HL. A phase 2 clinical trial assessing the efficacy and safety of pembrolizumab and radiotherapy in patients with metastatic triple-negative breast cancer. Cancer 2020; 126:850-860. [PMID: 31747077 DOI: 10.1002/cncr.32599] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/05/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The current study was conducted to evaluate the efficacy and safety of pembrolizumab-mediated programmed cell death protein 1 inhibition plus radiotherapy (RT) in patients with metastatic triple-negative breast cancer who were unselected for programmed death-ligand 1 expression. METHODS The current study was a single-arm, Simon 2-stage, phase 2 clinical trial that enrolled a total of 17 patients with a median age of 52 years (range, 37-73 years). An RT dose of 3000 centigrays (cGy) was delivered in 5 daily fractions. Pembrolizumab was administered intravenously at a dose of 200 mg within 3 days of the first RT fraction, and then every 3 weeks ± 3 days until disease progression. The median follow-up was 34.5 weeks (range, 2.1-108.3 weeks). The primary endpoint of the current study was the overall response rate (ORR) at week 13 in patients with unirradiated lesions measured using Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1). Secondary endpoints included safety and progression-free survival. Exploratory objectives were to identify biomarkers predictive of ORR and progression-free survival. RESULTS The ORR for the entire cohort was 17.6% (3 of 17 patients; 95% CI, 4.7%-44.2%), with 3 complete responses (CRs), 1 case of stable disease, and 13 cases of progressive disease. Eight patients died prior to week 13 due to disease progression. Among the 9 women assessed using RECIST version 1.1 at week 13, 3 (33%) achieved a CR, with a 100% reduction in tumor volume outside of the irradiated portal. The CRs were durable for 18 weeks, 20 weeks, and 108 weeks, respectively. The most common grade 1 to 2 toxicity (assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0) was dermatitis (29%). Four grade 3 adverse events were attributed to pembrolizumab: fatigue, lymphopenia, and infection. No were no grade 4 adverse events or treatment-related deaths reported. CONCLUSIONS The combination of pembrolizumab and RT was found to be safe and demonstrated encouraging activity in patients with poor-prognosis, metastatic, triple-negative breast cancer who were unselected for programmed death-ligand 1 expression. Larger clinical trials of checkpoint blockade plus RT with predictive biomarkers of response are needed.
Collapse
Affiliation(s)
- Alice Y Ho
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brittany B Arnold
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zishuo I Hu
- Medical Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ayca Gucalp
- Medical Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lizza Lebron-Zapata
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cindy Kallman
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Alessandro D'Agnolo
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samantha A Dunn
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Heather L McArthur
- Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
516
|
First-Line Treatment With Atezolizumab Plus Nab-Paclitaxel for Advanced Triple-Negative Breast Cancer. Am J Clin Oncol 2020; 43:340-348. [DOI: 10.1097/coc.0000000000000671] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
517
|
Shah AN, Flaum L, Helenowski I, Santa-Maria CA, Jain S, Rademaker A, Nelson V, Tsarwhas D, Cristofanilli M, Gradishar W. Phase II study of pembrolizumab and capecitabine for triple negative and hormone receptor-positive, HER2-negative endocrine-refractory metastatic breast cancer. J Immunother Cancer 2020; 8:e000173. [PMID: 32060053 PMCID: PMC7057426 DOI: 10.1136/jitc-2019-000173] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Response rates to single agent immune checkpoint blockade in unselected pretreated HER2-negative metastatic breast cancer (MBC) are low. However, they may be augmented when combined with chemotherapy. METHODS We conducted a single-arm, phase II study of patients with triple negative (TN) or hormone receptor-positive endocrine-refractory (HR+) MBC who were candidates for capecitabine. Patients were treated with pembrolizumab 200 mg intravenously day 1 and capecitabine 1000 mg/m2 by mouth twice daily on days 1-14 of a 21-day cycle. The primary end point was median progression-free survival (mPFS) compared with historic controls and secondary end points were overall response rate (ORR), safety and tolerability. The study had 80% power to detect a 2-month improvement in mPFS with the addition of pembrolizumab over historic controls treated with capecitabine alone. RESULTS Thirty patients, 16 TN and 14 HR+ MBC, were enrolled from 2017 to 2018. Patients had a median age of 51 years and received a median of 1 (range 0-6) prior lines of therapy for MBC. Of 29 evaluable patients, the mPFS was 4.0 (95% CI 2.0 to 6.4) months and was not significantly longer than historic controls of 3 months. The median overall survival was 15.4 (95% CI 8.2 to 20.3) months. The ORR was 14% (n=4), stable disease (SD) was 41% (n=12) and clinical benefit rate (CBR=partial response+SD>6 months) was 28% (n=8). The ORR and CBR were not significantly different between disease subtypes (ORR 13% and 14%, CBR 25% and 29% for TN and HR+, respectively). The 1-year PFS rate was 20.7% and three patients have ongoing responses. The most common adverse events were low grade and consistent with those seen in MBC patients receiving capecitabine, including hand-foot syndrome, gastrointestinal symptoms, fatigue and cytopenias. Toxicities at least possibly from pembrolizumab included grade 3 or 4 liver test abnormalities (7%), rash (7%) and diarrhea (3%), as well as grade 5 hepatic failure in a patient with liver metastases. CONCLUSIONS Compared with historical controls, pembrolizumab with capecitabine did not improve PFS in this biomarker unselected, pretreated cohort. However, some patients had prolonged disease control. TRIAL REGISTRATION NUMBER NCT03044730.
Collapse
Affiliation(s)
- Ami N Shah
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Lisa Flaum
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Irene Helenowski
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Cesar A Santa-Maria
- Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Sarika Jain
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Alfred Rademaker
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Valerie Nelson
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Dean Tsarwhas
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Massimo Cristofanilli
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - William Gradishar
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
518
|
Hutchinson KE, Yost SE, Chang CW, Johnson RM, Carr AR, McAdam PR, Halligan DL, Chang CC, Schmolze D, Liang J, Yuan Y. Comprehensive Profiling of Poor-Risk Paired Primary and Recurrent Triple-Negative Breast Cancers Reveals Immune Phenotype Shifts. Clin Cancer Res 2020; 26:657-668. [PMID: 31611282 PMCID: PMC8568263 DOI: 10.1158/1078-0432.ccr-19-1773] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/07/2019] [Accepted: 10/09/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Emerging data suggest immune checkpoint inhibitors have reduced efficacy in heavily pretreated triple-negative breast cancers (TNBC), but underlying mechanisms are poorly understood. To better understand the phenotypic evolution of TNBCs, we studied the genomic and transcriptomic profiles of paired tumors from patients with TNBC. EXPERIMENTAL DESIGN We collected paired primary and metastatic TNBC specimens from 43 patients and performed targeted exome sequencing and whole-transcriptome sequencing. From these efforts, we ascertained somatic mutation profiles, tumor mutational burden (TMB), TNBC molecular subtypes, and immune-related gene expression patterns. Stromal tumor-infiltrating lymphocytes (stromal TIL), recurrence-free survival, and overall survival were also analyzed. RESULTS We observed a typical TNBC mutational landscape with minimal shifts in copy number or TMB over time. However, there were notable TNBC molecular subtype shifts, including increases in the Lehmann/Pietenpol-defined basal-like 1 (BL1, 11.4%-22.6%) and mesenchymal (M, 11.4%-22.6%) phenotypes, and a decrease in the immunomodulatory phenotype (IM, 31.4%-3.2%). The Burstein-defined basal-like immune-activated phenotype was also decreased (BLIA, 42.2%-17.2%). Among downregulated genes from metastases, we saw enrichment of immune-related Kyoto Encyclopedia of Genes and Genomes pathways and gene ontology (GO) terms, and decreased expression of immunomodulatory gene signatures (P < 0.03) and percent stromal TILs (P = 0.03). There was no clear association between stromal TILs and survival. CONCLUSIONS We observed few mutational shifts, but largely consistent transcriptomic shifts in longitudinally paired TNBCs. Transcriptomic and IHC analyses revealed significantly reduced immune-activating gene expression signatures and TILs in recurrent TNBCs. These data may explain the observed lack of efficacy of immunotherapeutic agents in heavily pretreated TNBCs. Further studies are ongoing to better understand these initial observations.See related commentary by Savas and Loi, p. 526.
Collapse
Affiliation(s)
| | - Susan E Yost
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, California
| | - Ching-Wei Chang
- Oncology Biostatistics, Genentech, Inc., South San Francisco, California
| | | | | | | | | | - Chun-Chieh Chang
- Oncology Biostatistics, Genentech, Inc., South San Francisco, California
| | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, California
| | - Jackson Liang
- Oncology Biomarker Development, Genentech, Inc., South San Francisco, California
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, California.
| |
Collapse
|
519
|
Yeong J, Tan T, Chow ZL, Cheng Q, Lee B, Seet A, Lim JX, Lim JCT, Ong CCH, Thike AA, Saraf S, Tan BYC, Poh YC, Yee S, Liu J, Lim E, Iqbal J, Dent R, Tan PH. Multiplex immunohistochemistry/immunofluorescence (mIHC/IF) for PD-L1 testing in triple-negative breast cancer: a translational assay compared with conventional IHC. J Clin Pathol 2020; 73:557-562. [PMID: 31969377 DOI: 10.1136/jclinpath-2019-206252] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) monoclonal antibody therapy has recently gained approval for treating metastatic triple-negative breast cancer (TNBC) -, in particular in the PD-L1+ patient subgroup of the recent IMpassion130 trial. The SP142 PD-L1 antibody clone was used as a predictive assay in this trial, but this clone was found to be an outlier in previous harmonisation studies in lung cancer. AIMS To address the comparability of PD-L1 clones in TNBC, we evaluated the concordance between conventional immunohistochemistry (IHC) and multiplex immunohistochemistry/immunofluorescence (mIHC/IF) that allowed simultaneous quantification of three different PD-L1 antibodies (22C3, SP142 and SP263). METHODS Our cohort comprised 25 TNBC cases, 12 non-small-cell lung carcinomas and 8 other cancers. EpCAM labelling was used to distinguish tumour cells from immune cells. RESULTS Moderate-to-strong correlations in PD-L1 positivity were found between results obtained through mIHC/IF and IHC. Individual concordance rates in the study ranged from 67% to 100%, with Spearman's rank correlation coefficient values up to 0.88. CONCLUSIONS mIHC/IF represents a promising tool in the era of cancer immunotherapy, as it can simultaneously detect and quantify PD-L1 labelling with multiple antibody clones, and allow accurate evaluation of tumour and immune cells. Clinicians and pathologists require this information to predict patient response to anti-PD-1/PD-L1 therapy. The adoption of this assay may represent a significant advance in the management of therapeutically challenging cancers. Further analysis and assay harmonisation are essential for translation to a routine diagnostic setting.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore .,Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Tira Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Zi Long Chow
- Division of Pathology, Singapore General Hospital, Singapore.,University of Tasmania, Hobart, Tasmania, Australia
| | - Qing Cheng
- Duke-NUS Medical School, Duke-NUS Medical School, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Amanda Seet
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | | | - Jeffrey Chun Tatt Lim
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Clara Chong Hui Ong
- Division of Pathology, Singapore General Hospital, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, Singapore
| | - Sahil Saraf
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Yong Cheng Poh
- Diagnostics Development (DxD) Hub, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Sidney Yee
- Diagnostics Development (DxD) Hub, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Jin Liu
- Duke-NUS Medical School, Duke-NUS Medical School, Singapore
| | - Elaine Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, Singapore
| | - Rebecca Dent
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| |
Collapse
|
520
|
Morgan E, Suresh A, Ganju A, Stover DG, Wesolowski R, Sardesai S, Noonan A, Reinbolt R, VanDeusen J, Williams N, Cherian MA, Li Z, Young G, Palettas M, Stephens J, Liu J, Luff A, Ramaswamy B, Lustberg M. Assessment of outcomes and novel immune biomarkers in metaplastic breast cancer: a single institution retrospective study. World J Surg Oncol 2020; 18:11. [PMID: 31937323 PMCID: PMC6961248 DOI: 10.1186/s12957-019-1780-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/29/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Metaplastic breast cancer remains poorly characterized given its rarity and heterogeneity. The majority of metaplastic breast cancers demonstrate a phenotype of triple-negative breast cancer; however, differences in clinical outcomes between metaplastic breast cancer and triple-negative breast cancer in the era of third-generation chemotherapy remain unclear. METHODS We compared the clinical outcomes between women with metaplastic breast cancer and women with triple-negative breast cancer diagnosed between 1994 and 2014. Metaplastic breast cancer patients were matched 1:3 to triple-negative breast cancer patients by stage and age at diagnosis. Distant disease-free survival (DDFS) and overall survival (OS) were estimated using Kaplan Meier methods and Cox proportional hazard regression models. Immune checkpoint markers were characterized by immunohistochemistry in a subset of samples. RESULTS Forty-four metaplastic breast cancer patients (stage I 14%; stage II 73%; stage III 11%; stage IV 2%) with an average age of 55.4 (± 13.9) years at diagnosis. Median follow-up for the included metaplastic breast cancer and triple-negative breast cancer patients (n = 174) was 2.8 (0.1-19.0) years. The DDFS and OS between matched metaplastic breast cancer and triple-negative breast cancer patients were similar, even when adjusting for clinical covariates (DDFS: HR = 1.64, p = 0.22; OS: HR = 1.64, p = 0.26). Metaplastic breast cancer samples (n = 27) demonstrated greater amount of CD163 in the stroma (p = 0.05) and PD-L1 in the tumor (p = 0.01) than triple-negative breast cancer samples (n = 119), although more triple-negative breast cancer samples were positive for CD8 in the tumor than metaplastic breast cancer samples (p = 0.02). CONCLUSIONS Patients with metaplastic breast cancer had similar outcomes to those with triple-negative breast cancer based on DDFS and OS. The immune checkpoint marker profile of metaplastic breast cancers in this study may prove useful in future studies attempting to demonstrate an association between immune profile and survival.
Collapse
MESH Headings
- B7-H1 Antigen/immunology
- Biomarkers, Tumor/immunology
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/therapy
- Combined Modality Therapy
- Female
- Follow-Up Studies
- Humans
- Metaplasia/pathology
- Metaplasia/therapy
- Middle Aged
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Neoplasm Staging
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Retrospective Studies
- Survival Rate
- Triple Negative Breast Neoplasms/immunology
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/therapy
Collapse
Affiliation(s)
- Evan Morgan
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Anupama Suresh
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Akaansha Ganju
- Department of Internal Medicine, Riverside Methodist Hospital, Columbus, Ohio USA
| | - Daniel G. Stover
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Robert Wesolowski
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Sagar Sardesai
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Anne Noonan
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Raquel Reinbolt
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Jeffrey VanDeusen
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Nicole Williams
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Mathew A. Cherian
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Zaibo Li
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH USA
| | - Gregory Young
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH USA
| | - Marilly Palettas
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH USA
| | - Julie Stephens
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH USA
| | - Joseph Liu
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Amanda Luff
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Bhuvaneswari Ramaswamy
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| | - Maryam Lustberg
- Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH USA
- Division of Medical Oncology, Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, OH USA
| |
Collapse
|
521
|
Li W, Qie J, Zhang Y, Chang J. Spatiotemporal Changes in Checkpoint Molecule Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:167-200. [PMID: 32185711 DOI: 10.1007/978-981-15-3266-5_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 blockade, have led to therapeutic breakthrough in patients with advanced malignancy, covering the lung, breast, gastrointestinal, head and neck, urinary system, lymphoma, and solid tumor harboring MSI/dMMR. In certain cancer types, the expression level of immune checkpoint molecule will be required if the immune-based approaches are considered, especially the PD-L1 expression. However, in other types, survival benefit has been proven regardless of PD-L1 expression. It raises a question of how to select patients for immune therapy and whether the expression of immune checkpoint molecules will be optimal biomarkers. Before answering this question, a comprehensive map for the expression of immune checkpoint molecules is needed. In this chapter, we describe our current knowledge on the spatiotemporal changes in the expression of checkpoint molecules. We discuss the different frequencies of expression depending on tumor types and stages, the different patterns between primary and metastatic tumors, as well as the change of expression before and after treatment. The expression of PD-L1 has been most studied, but the threshold that separate "positive" and "negative" PD-L1 expressions and the consistency of testing platform remain under debate. Better understanding on the tumor microenvironment and expression of checkpoint molecules will help to identify patients who will benefit from checkpoint blockade therapy.
Collapse
Affiliation(s)
- Wenhua Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Jingbo Qie
- Institutes of Biomedical Sciences, Fudan University, 130 Dongan Road, Shanghai, 200032, China
| | - Yao Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jinjia Chang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| |
Collapse
|
522
|
Elia G, Ferrari SM, Galdiero MR, Ragusa F, Paparo SR, Ruffilli I, Varricchi G, Fallahi P, Antonelli A. New insight in endocrine-related adverse events associated to immune checkpoint blockade. Best Pract Res Clin Endocrinol Metab 2020; 34:101370. [PMID: 31983543 DOI: 10.1016/j.beem.2019.101370] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Anticancer immunotherapy, in the form of immune checkpoint inhibition, is a paradigm shift that has transformed the care of patients with different types of solid and hematologic cancers. The most notable improvements have been seen in patients with melanoma, non-small-cell lung, bladder, renal, cervical, urotherial, and colorectal cancers, Merkel cell carcinoma, and Hodgkin lymphoma. Monoclonal antibodies (mAbs) targeting immune checkpoints (i.e., anti-CTLA: ipilimumab; anti-PD-1: nivolumab, pembrolizumab; anti-PD-L1: durvalumab, atezolizumab, avelumab) unleash the immune system against tumor cells targeting mainly T cells. Treatment with immune checkpoint inhibitors (ICIs) is associated with a variety of diverse and distinct immune-related adverse events (irAEs), reflecting the mechanistic underpinning of each target (i.e., CTLA-4, and PD-1/PD-L1 network). The most frequent endocrine irAEs associated with anti-PD-1 mAb treatment are thyroid dysfunctions, whereas hypophysitis is mostly linked to anti-CTLA-4 treatment. Type 1 diabetes mellitus and adrenalitis are rare irAEs. Combination therapy (anti-CTLA-4 plus anti-PD-1/PD-L1) can be associated with an increased risk and prevalence of endocrine irAEs. In this paper we discuss the pathophysiological and clinical aspects of irAEs with specific emphasis on endocrine irAEs associated with ICIs. With a growing number of patients treated with ICIs, a tight collaboration among oncologists, endocrinologists and immunologists appears necessary when the circumstances are more challenging and for better management of severe endocrine irAEs. Further investigations are urgently needed to better understand the mechanisms by which different ICIs can induce a variety of endocrine irAEs.
Collapse
Affiliation(s)
- Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy.
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy.
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; WAO Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy.
| | - Sabrina Rosaria Paparo
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy.
| | - Ilaria Ruffilli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy.
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; WAO Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Poupak Fallahi
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, 56126, Pisa, Italy.
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy.
| |
Collapse
|
523
|
Jang BS, Han W, Kim IA. Tumor mutation burden, immune checkpoint crosstalk and radiosensitivity in single-cell RNA sequencing data of breast cancer. Radiother Oncol 2020; 142:202-209. [DOI: 10.1016/j.radonc.2019.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/23/2019] [Accepted: 11/04/2019] [Indexed: 02/04/2023]
|
524
|
Mina LA, Lim S, Bahadur SW, Firoz AT. Immunotherapy for the Treatment of Breast Cancer: Emerging New Data. BREAST CANCER (DOVE MEDICAL PRESS) 2019; 11:321-328. [PMID: 32099454 PMCID: PMC6997226 DOI: 10.2147/bctt.s184710] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
Breast cancer is the most common type of cancer affecting women in the United States. Triple-negative breast cancer remains the most aggressive molecular subtype secondary to a lack of therapeutic targets. The search for a target has led us to investigate immunotherapeutic agents. Immunotherapy has recently demonstrated significant breakthroughs in various types of cancers that are refractory to traditional therapies including melanoma and Non-Small Cell Lung Cancer (NSCLC). Breast cancer however remains one of the tumors that was initially least investigated because of being considered to have a low immunogenic potential and a low mutational load. Over the past few years, antiPD1/PDL1 drugs have started to make progress in the triple-negative subtype with more promising outcomes. In this report, we review the treatment of triple-negative breast cancer and specifically shed light on advances in immunotherapy and newly approved drugs in this challenging disease.
Collapse
Affiliation(s)
- Lida A Mina
- Hematology Oncology Department, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Shannon Lim
- Pharmacy Department, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Shakeela W Bahadur
- Hematology Oncology Department, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Abdul T Firoz
- Science Department, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
525
|
Iwata H, Inoue K, Kaneko K, Ito Y, Tsugawa K, Hasegawa A, Nakagawa S, Kuratomi H, Tamura K. Subgroup analysis of Japanese patients in a Phase 3 study of atezolizumab in advanced triple-negative breast cancer (IMpassion130). Jpn J Clin Oncol 2019; 49:1083-1091. [PMID: 31612909 PMCID: PMC6935297 DOI: 10.1093/jjco/hyz135] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In the randomised Phase 3 IMpassion130 trial, atezolizumab combined with nab-paclitaxel (atezo + nab-P) in 902 patients with triple-negative breast cancer (TNBC) showed prolonged progression-free survival (PFS) in both the intention-to-treat (ITT) population and programmed death-ligand 1 (PD-L1)-positive subgroup compared with placebo plus nab-P (plac + nab-P). This study assessed the efficacy and safety of atezo + nab-P in the IMpassion130 Japanese subpopulation. METHODS Eligible patients had unresectable locally advanced or metastatic TNBC previously untreated with chemotherapy for metastatic disease. Patients were randomised 1:1 to receive either atezo + nab-P or plac + nab-P. Co-primary endpoints were investigator-assessed PFS and overall survival (ITT population and PD-L1-positive subgroup). These were also assessed in the Japanese subpopulation. RESULTS There were 65 Japanese patients (34 atezo + nab-P; 31 plac + nab-P). The PD-L1-positive subgroup included 25 patients (12 atezo + nab-P; 13 plac + nab-P). Median PFS was 7.4 months (atezo + nab-P) versus 4.6 months (plac + nab-P; hazard ratio [HR], 0.47; 95% CI, 0.25-0.90). In the PD-L1-positive subgroup, median PFS was 10.8 months (atezo + nab-P) versus 3.8 months (plac + nab-P; HR, 0.04; 95% CI, <0.01-0.35). Safety results in the Japanese subgroup were consistent with those in the overall population. The Japanese subgroup had a lower incidence of adverse events leading to treatment withdrawal than the overall population. More patients in the atezo + nab-P arm had neutrophil count decreases and stomatitis than patients in the plac + nab-P arm. CONCLUSIONS Atezo + nab-P efficacy in Japanese patients was consistent with the overall IMpassion130 population. No new safety signals were observed, and tolerability was consistent with that of the overall population.
Collapse
Affiliation(s)
- Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center, Aichi, Japan
| | - Kenichi Inoue
- Division of Breast Oncology, Saitama Cancer Center, Saitama, Japan
| | - Koji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yoshinori Ito
- Breast Medical Oncology, The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koichiro Tsugawa
- Division of Breast and Endocrine Surgery, Department of Surgery, St Marianna University School of Medicine Hospital, Kanagawa, Japan
| | | | | | | | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
526
|
Nederlof I, De Bortoli D, Bareche Y, Nguyen B, de Maaker M, Hooijer GKJ, Buisseret L, Kok M, Smid M, Van den Eynden GGGM, Brinkman AB, Hudecek J, Koster J, Sotiriou C, Larsimont D, Martens JWM, van de Vijver MJ, Horlings HM, Salgado R, Biganzoli E, Desmedt C. Comprehensive evaluation of methods to assess overall and cell-specific immune infiltrates in breast cancer. Breast Cancer Res 2019; 21:151. [PMID: 31878981 PMCID: PMC6933637 DOI: 10.1186/s13058-019-1239-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/16/2019] [Indexed: 12/23/2022] Open
Abstract
Background Breast cancer (BC) immune infiltrates play a critical role in tumor progression and response to treatment. Besides stromal tumor infiltrating lymphocytes (sTILs) which have recently reached level 1B evidence as a prognostic marker in triple negative BC, a plethora of methods to assess immune infiltration exists, and it is unclear how these compare to each other and if they can be used interchangeably. Methods Two experienced pathologists scored sTIL, intra-tumoral TIL (itTIL), and 6 immune cell types (CD3+, CD4+, CD8+, CD20+, CD68+, FOXP3+) in the International Cancer Genomics Consortium breast cancer cohort using hematoxylin and eosin-stained (n = 243) and immunohistochemistry-stained tissue microarrays (n = 254) and whole slides (n = 82). The same traits were evaluated using transcriptomic- and methylomic-based deconvolution methods or signatures. Results The concordance correlation coefficient (CCC) between pathologists for sTIL was very good (0.84) and for cell-specific immune infiltrates slightly lower (0.63–0.66). Comparison between tissue microarray and whole slide pathology scores revealed systematically higher values in whole slides (ratio 2.60–5.98). The Spearman correlations between microscopic sTIL and transcriptomic- or methylomic-based assessment of immune infiltrates were highly variable (r = 0.01–0.56). Similar observations were made for cell type-specific quantifications (r = 0.001–0.54). We observed a strong inter-method variability between the omics-derived estimations, which is further cell type dependent. Finally, we demonstrated that most methods more accurately identify highly infiltrated (sTIL ≥ 60%; area under the curve, AUC, 0.64–0.99) as compared to lowly infiltrated tumors (sTIL ≤ 10%; AUC 0.52–0.82). Conclusions There is a lower inter-pathologist concordance for cell-specific quantification as compared to overall infiltration quantification. Microscopic assessments are underestimated when considering small cores (tissue microarray) instead of whole slides. Results further highlight considerable differences between the microscopic-, transcriptomic-, and methylomic-based methods in the assessment of overall and cell-specific immune infiltration in BC. We therefore call for extreme caution when assessing immune infiltrates using current methods and emphasize the need for standardized immune characterization beyond TIL.
Collapse
Affiliation(s)
- Iris Nederlof
- Department of Pathology, Amsterdam University Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Davide De Bortoli
- Unit of Medical Statistics, Biometry and Bioinformatics "Giulio A. Maccacaro," Department of Clinical Sciences and Community Health and DSRC, University of Milan, Campus Cascina Rosa, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Yacine Bareche
- J.C. Heuson Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, 1000, Brussels, Belgium
| | - Bastien Nguyen
- J.C. Heuson Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, 1000, Brussels, Belgium
| | - Michiel de Maaker
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Gerrit K J Hooijer
- Department of Pathology, Amsterdam University Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Laurence Buisseret
- J.C. Heuson Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, 1000, Brussels, Belgium
| | - Marleen Kok
- Departments of Medical Oncology and Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marcel Smid
- Department of Medical Oncology and Cancer Genomics Centre Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | | | - Arie B Brinkman
- Department of Molecular Biology, Nijmegen Centre for Molecular Life Sciences, Faculty of Science, Radboud University, 6500 HB, Nijmegen, The Netherlands
| | - Jan Hudecek
- Department of Research IT, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jan Koster
- Department of Oncogenomics, Amsterdam University Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christos Sotiriou
- J.C. Heuson Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles, Institut Jules Bordet, 1000, Brussels, Belgium
| | - Denis Larsimont
- Pathology Department, Institut Jules Bordet, 1000, Brussels, Belgium
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Centre Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Marc J van de Vijver
- Department of Pathology, Amsterdam University Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Hugo M Horlings
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Ziekenhuizen, Wilrijk, Belgium.,Division of Research, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Elia Biganzoli
- Unit of Medical Statistics, Biometry and Bioinformatics "Giulio A. Maccacaro," Department of Clinical Sciences and Community Health and DSRC, University of Milan, Campus Cascina Rosa, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Christine Desmedt
- Department of Oncology, Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
527
|
Vranic S, Cyprian FS, Gatalica Z, Palazzo J. PD-L1 status in breast cancer: Current view and perspectives. Semin Cancer Biol 2019; 72:146-154. [PMID: 31883913 DOI: 10.1016/j.semcancer.2019.12.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022]
Abstract
Breast cancer was traditionally not considered a particularly immunogenic tumor. However, recent developments have shown that some aggressive triple-negative breast cancers are immunogenic, exhibit a resistance to chemotherapy and have a poor prognosis. These cancers have been shown to express molecules identified as targets for immunotherapy. Despite the advances, the challenges are many, and include identifying the patients that may benefit from immunotherapy. The best methods to analyze these samples and to evaluate immunogenicity are also major challenges. Therefore, the most accurate and reliable assessment of immune cells as potential targets is one of the most important aims in the current research in breast immunotherapy. In the present review, we briefly discuss the mechanisms of the regulation of checkpoint inhibitors (PD-1/PD-L1) in breast cancer and explore the predictive aspects in the PD-L1 testing.
Collapse
Affiliation(s)
- Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | | | | |
Collapse
|
528
|
Azim HA, Ghosn M, Oualla K, Kassem L. Personalized treatment in metastatic triple-negative breast cancer: The outlook in 2020. Breast J 2019; 26:69-80. [PMID: 31872557 DOI: 10.1111/tbj.13713] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022]
Abstract
Compared with other breast cancer subtypes, patients with triple-negative breast cancer (TNBC), and irrespective to their disease stage, were always recognized to have the worst overall survival data. Although this does not seem different at the present time, yet the last few years have witnessed many breakthrough genomic and molecular findings, that could dramatically improve our understanding of the biological complexity of TNBC. Based on genomic analyses, it was consistently evident that TNBC comprises a heterogeneous group of cancers, which have numerous diverse molecular aberrations. This-in return-has provided a platform for a new generation of clinical trials using many innovative therapies, directed against such novel targets. At the present time, two PARP inhibitors and one anti-PD-L1 monoclonal antibody (in combination with chemotherapy) have been approved in certain subpopulations of metastatic TNBC (mTNBC) patients, which have finally brought this disease into the era of personalized medicine. In the current review, we will explore the genomic landscape of TNBC, through which many actionable targets were graduated. We will also discuss the results of the key-practice changing-clinical studies, and some upcoming personalized treatment options for patients with mTNBC, that may be clinically adopted in the near future.
Collapse
Affiliation(s)
- Hamdy A Azim
- Clinical Oncology Department, Kasr Alainy School of Medicine, Cairo University, Giza, Egypt.,Clinical Oncology Department, Cairo Oncology Center, Cairo, Egypt
| | - Marwan Ghosn
- Hotel Dieu de France University Hospital and Saint Joseph University, Beirut, Lebanon
| | - Karima Oualla
- Medical Oncology Department, Hassan II University Hospital, Fes, Morocco
| | - Loay Kassem
- Clinical Oncology Department, Kasr Alainy School of Medicine, Cairo University, Giza, Egypt.,Clinical Oncology Department, Cairo Oncology Center, Cairo, Egypt
| |
Collapse
|
529
|
Tian Y, Wang X, Zhao S, Liao X, Younis MR, Wang S, Zhang C, Lu G. JQ1-Loaded Polydopamine Nanoplatform Inhibits c-MYC/Programmed Cell Death Ligand 1 to Enhance Photothermal Therapy for Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46626-46636. [PMID: 31751121 DOI: 10.1021/acsami.9b18730] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Programmed cell death ligand 1 (PD-L1) blockade has achieved great success in cancer immunotherapy; however, the response of triple-negative breast cancer (TNBC) to PD-L1 antibodies is limited. To address this challenge, we use the bromodomain and extra-terminal inhibitor JQ1 to down-regulate the expression of PD-L1 and thus elicit the immune response to TNBC instead of using antibodies to block PD-L1. JQ1 also inhibits the growth of TNBC as a targeted therapeutic agent by inhibiting the BRD4-c-MYC axis. The polydopamine nanoparticles (PDMNs) are introduced as a biodegradable and adaptable platform to load JQ1 and induce photothermal therapy (PTT) as another synergistic therapeutic modality. Because the JQ1-loaded PDMNs (PDMN-JQ1) are self-degradable and release JQ1 continuously, this synergistic treatment can lead to remarkable activation of cytotoxic T lymphocytes and induce a strong immune-memory effect to protect mice from tumor re-challenge. Taken together, our study demonstrates a compact and simple nanoplatform for triple therapy, including targeted therapy, PTT, and immunotherapy, for TNBC treatment.
Collapse
Affiliation(s)
| | | | | | | | - Muhammad Rizwan Younis
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210093 , Jiangsu , P.R. China
| | - Shouju Wang
- Department of Radiology , First Affiliated Hospital of Nanjing Medical University , Nanjing 210029 , Jiangsu , P.R. China
| | | | - Guangming Lu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210093 , Jiangsu , P.R. China
| |
Collapse
|
530
|
Li Q, Wang Y, Jia W, Deng H, Li G, Deng W, Chen J, Kim BYS, Jiang W, Liu Q, Liu J. Low-Dose Anti-Angiogenic Therapy Sensitizes Breast Cancer to PD-1 Blockade. Clin Cancer Res 2019; 26:1712-1724. [PMID: 31848190 DOI: 10.1158/1078-0432.ccr-19-2179] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/03/2019] [Accepted: 12/12/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite its enormous successes, the overall response rate of cancer immunotherapy remains suboptimal, especially in breast cancer. There is an increased interest in combining immune checkpoint inhibitor with targeted agents to enhance antitumor effect. Anti-angiogenic drugs have been shown to synergize with immune checkpoint blockades, but the optimal setting for combining these two modalities and the underlying mechanisms of synergistic responses are not fully understood. EXPERIMENTAL DESIGN We tested the combination of anti-PD-1 and different doses of VEGFR2-targeting agents in syngeneic breast cancer mouse models. Tumor-infiltrated immune cell subsets were profiled by flow cytometry. A cytokine array was carried out to identify inflammatory changes in different treatment conditions. The efficacy of combined anti-angiogenic and anti-PD-1 therapy was further evaluated in patients with advanced triple-negative breast cancer (TNBC). RESULTS Blockade of VEGFR2 sensitizes breast tumors to PD-1 blockade in a dose-dependent manner. Although both conventional and low-dose anti-VEGFR2 antibody treatments normalize tumor vessels, low-dose VEGFR2 blockade results in more robust immune cell infiltration and activation and promotes the secretion of osteopontin (OPN) by CD8+ T cells. OPN subsequently induces tumor cell production of TGF-β, which in turn upregulates PD-1 expression on immune cells. In patients with advanced TNBC, combined treatment with low-dose anti-VEGFR2 inhibitor and anti-PD-1 demonstrated excellent tolerability and efficacy. Higher OPN and TGF-β expressions correlated with improved treatment responses. CONCLUSIONS Together, these results demonstrate a dose-dependent synergism between anti-angiogenic therapy and immune checkpoint blockade, thus providing important insights into the optimal strategies for combining immunotherapy with molecular-targeted agents.
Collapse
Affiliation(s)
- Qian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Weijuan Jia
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Heran Deng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangdi Li
- Department of Public Health, Central South University, Changsha, China
| | - Weiye Deng
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jiewen Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China. .,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieqiong Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China. .,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
531
|
Page DB, Pucilowska J, Sanchez KG, Conrad VK, Conlin AK, Acheson AK, Perlewitz KS, Imatani JH, Aliabadi-Wahle S, Moxon N, Mellinger SL, Seino AY, Martel M, Wu Y, Sun Z, Redmond WL, Rajamanickam V, Waddell D, Laxague D, Shah M, Chang SC, Urba WJ. A Phase Ib Study of Preoperative, Locoregional IRX-2 Cytokine Immunotherapy to Prime Immune Responses in Patients with Early-Stage Breast Cancer. Clin Cancer Res 2019; 26:1595-1605. [PMID: 31831558 DOI: 10.1158/1078-0432.ccr-19-1119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE To evaluate the safety and feasibility of preoperative locoregional cytokine therapy (IRX-2 regimen) in early-stage breast cancer, and to evaluate for intratumoral and peripheral immunomodulatory activity. PATIENTS AND METHODS Sixteen patients with stage I-III early-stage breast cancer (any histology type) indicated for surgical lumpectomy or mastectomy were enrolled to receive preoperative locoregional immunotherapy with the IRX-2 cytokine biological (2 mL subcutaneous × 10 days to periareolar skin). The regimen also included single-dose cyclophosphamide (300 mg/m2) on day 1 to deplete T-regulatory cells and oral indomethacin to modulate suppressive myeloid subpopulations. The primary objective was to evaluate feasibility (i.e., receipt of therapy without surgical delays or grade 3/4 treatment-related adverse events). The secondary objective was to evaluate changes in stromal tumor-infiltrating lymphocyte score. The exploratory objective was to identify candidate pharmacodynamic changes for future study using a variety of assays, including flow cytometry, RNA and T-cell receptor DNA sequencing, and multispectral immunofluorescence. RESULTS Preoperative locoregional cytokine administration was feasible in 100% (n = 16/16) of subjects and associated with increases in stromal tumor-infiltrating lymphocytes (P < 0.001). Programmed death ligand 1 (CD274) was upregulated at the RNA (P < 0.01) and protein level [by Ventana PD-L1 (SP142) and immunofluorescence]. Other immunomodulatory effects included upregulation of RNA signatures of T-cell activation and recruitment and cyclophosphamide-related peripheral T-regulatory cell depletion. CONCLUSIONS IRX-2 is safe in early-stage breast cancer. Potentially favorable immunomodulatory changes were observed, supporting further study of IRX-2 in early-stage breast cancer and other malignancies.
Collapse
Affiliation(s)
- David B Page
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon.
| | - Joanna Pucilowska
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Katherine G Sanchez
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Valerie K Conrad
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Alison K Conlin
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Anupama K Acheson
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Kelly S Perlewitz
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - James H Imatani
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | | | - Nicole Moxon
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Staci L Mellinger
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Amanda Y Seino
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Martiza Martel
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Yaping Wu
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Zhaoyu Sun
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | | | - Dottie Waddell
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Deborah Laxague
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Monil Shah
- Brooklyn Therapeutics, Brooklyn, New York
| | - Shu-Ching Chang
- Medical Data Research Center, Providence St. Joseph Health, Portland, Oregon
| | - Walter J Urba
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| |
Collapse
|
532
|
Mavratzas A, Seitz J, Smetanay K, Schneeweiss A, Jäger D, Fremd C. Atezolizumab for use in PD-L1-positive unresectable, locally advanced or metastatic triple-negative breast cancer. Future Oncol 2019; 16:4439-4453. [PMID: 31829043 DOI: 10.2217/fon-2019-0468] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Since the US FDA-approval of the first immune checkpoint inhibitor, anticytotoxic T-lymphocyte antigen-4 monoclonal antibody ipilimumab, for metastatic melanoma on 28 March 2011, another six agents have been granted use among a multitude of tumors, including renal cell cancer, Hodgkin lymphoma, urothelial carcinoma and non-small-cell lung cancer. The first anti-programmed cell death ligand-1 monoclonal antibody to receive the FDA approval, atezolizumab (Tecentriq®), has yielded promising results among international Phase III trials in triple-negative breast cancer and small-cell lung cancer, expanding the field of cancer immunotherapies. Herein, we review the pharmacodynamic and pharmacokinetic properties of atezolizumab, its safety and efficacy data from early clinical trials and summarize data from Phase III IMpassion130 trial, prompting FDA and EMA approval of atezolizumab in metastatic triple-negative breast cancer. Finally, implications for clinical use and ongoing research will be briefly discussed.
Collapse
Affiliation(s)
- Athanasios Mavratzas
- National Center for Tumor Disease, Gynecologic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Julia Seitz
- National Center for Tumor Disease, Gynecologic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina Smetanay
- National Center for Tumor Disease, Gynecologic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Disease, Gynecologic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dirk Jäger
- National Center for Tumor Disease, Gynecologic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carlo Fremd
- National Center for Tumor Disease, Gynecologic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
533
|
Dowling RJO, Sparano JA, Goodwin PJ, Bidard FC, Cescon DW, Chandarlapaty S, Deasy JO, Dowsett M, Gray RJ, Henry NL, Meric-Bernstam F, Perlmutter J, Sledge GW, Thorat MA, Bratman SV, Carey LA, Chang MC, DeMichele A, Ennis M, Jerzak KJ, Korde LA, Lohmann AE, Mamounas EP, Parulekar WR, Regan MM, Schramek D, Stambolic V, Whelan TJ, Wolff AC, Woodgett JR, Kalinsky K, Hayes DF. Toronto Workshop on Late Recurrence in Estrogen Receptor-Positive Breast Cancer: Part 2: Approaches to Predict and Identify Late Recurrence, Research Directions. JNCI Cancer Spectr 2019; 3:pkz049. [PMID: 32337478 PMCID: PMC7050024 DOI: 10.1093/jncics/pkz049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/18/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Late disease recurrence (more than 5 years after initial diagnosis) represents a clinical challenge in the treatment and management of estrogen receptor-positive breast cancer (BC). An international workshop was convened in Toronto, Canada, in February 2018 to review the current understanding of late recurrence and to identify critical issues that require future study. The underlying biological causes of late recurrence are complex, with the processes governing cancer cell dormancy, including immunosurveillance, cell proliferation, angiogenesis, and cellular stemness, being integral to disease progression. These critical processes are described herein as well as their role in influencing risk of recurrence. Moreover, observational and interventional clinical trials are proposed, with a focus on methods to identify patients at risk of recurrence and possible strategies to combat this in patients with estrogen receptor-positive BC. Because the problem of late BC recurrence of great importance, recent advances in disease detection and patient monitoring should be incorporated into novel clinical trials to evaluate approaches to enhance patient management. Indeed, future research on these issues is planned and will offer new options for effective late recurrence treatment and prevention strategies.
Collapse
Affiliation(s)
- Ryan J O Dowling
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Joseph A Sparano
- Departments of Medicine and Medical Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Albert Einstein Cancer Center, New York, NY
| | - Pamela J Goodwin
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill-Cornell Medical College, New York, NY
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, The Royal Marsden NHS Foundation Trust, Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
| | - Robert J Gray
- Department of Biostatistics, Dana-Farber Cancer Institute, Boston, MA
- Harvard T.H. Chan School of Public Health, Boston, MA
| | - N Lynn Henry
- University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, Salt Lake City, UT
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - George W Sledge
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Mangesh A Thorat
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Scott V Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Martin C Chang
- University of Vermont Medical Center, Larner College of Medicine, Burlington, VT
| | - Angela DeMichele
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | - Katarzyna J Jerzak
- Division of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Larissa A Korde
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Ana Elisa Lohmann
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Wendy R Parulekar
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Meredith M Regan
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Vuk Stambolic
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Timothy J Whelan
- McMaster University and Juravinski Cancer Centre, Hamilton, ON, Canada
| | - Antonio C Wolff
- The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Jim R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Kevin Kalinsky
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Daniel F Hayes
- University of Michigan Rogel Cancer Center, and Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
534
|
Lu J, Li L, Lan Y, Liang Y, Meng H. Immune checkpoint inhibitor-associated pituitary-adrenal dysfunction: A systematic review and meta-analysis. Cancer Med 2019; 8:7503-7515. [PMID: 31679184 PMCID: PMC6912062 DOI: 10.1002/cam4.2661] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/25/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
With the growing use of immune checkpoint inhibitors (ICIs), case reports of rare yet life-threatening pituitary-adrenal dysfunctions, particularly for hypopituitarism, are increasingly being published. In this analysis, we focus on these events by including the most recent publications and reports from early phase I/II and phase III clinical trials and comparing the incidence and risks across different ICI regimens. PubMed, Embase, and the Cochrane Library were systematically searched from inception to April 2019 for clinical trials that reported on pituitary-adrenal dysfunction. The rates of events, odds ratios (ORs), and 95% confidence intervals (CIs) were obtained using random effects meta-analysis. The analyses included data from 160 trials involving 40 432 participants. The rate was 2.43% (95% CI, 1.73%-3.22%) for all-grade adrenal insufficiency and 3.25% (95% CI, 2.15%-4.51%) for hypophysitis. Compared with the placebo or other therapeutic regimens, ICI agents were associated with a higher incidence of serious-grade adrenal insufficiency (OR 3.19, 95% CI, 1.84 to 5.54) and hypophysitis (OR 4.77, 95% CI, 2.60 to 8.78). Among 71 serious-grade hypopituitarism instances in 12 336 patients, there was a significant association between ICIs and hypopituitarism (OR 3.62, 95% CI, 1.86 to 7.03). Substantial heterogeneity was noted across the studies for the rates of these events, which in part was attributable to the different types of ICIs and varied phases of the clinical trials. Although the rates of these events were low, the risk was increased following ICI-based treatment, particularly for CTLA-4 inhibitors, which were associated with a higher incidence of pituitary-adrenal dysfunction than PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Jingli Lu
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Lulu Li
- Department of PharmacyWuhan No.1 HospitalWuhanHubeiChina
| | - Yan Lan
- Department of PharmacyHuangshi Center HospitalHuangshiHubeiChina
| | - Yan Liang
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Haiyang Meng
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
535
|
Ascierto PA, Bifulco C, Buonaguro L, Emens LA, Ferris RL, Fox BA, Delgoffe GM, Galon J, Gridelli C, Merlano M, Nathan P, Odunsi K, Okada H, Paulos CM, Pignata S, Schalper KA, Spranger S, Tortora G, Zarour H, Butterfield LH, Puzanov I. Perspectives in immunotherapy: meeting report from the "Immunotherapy Bridge 2018" (28-29 November, 2018, Naples, Italy). J Immunother Cancer 2019; 7:332. [PMID: 31783779 PMCID: PMC6884742 DOI: 10.1186/s40425-019-0798-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is now widely established as a potent and effective treatment option across several types of cancer. However, there is increasing recognition that not all patients respond to immunotherapy, focusing attention on the immune contexture of the tumor microenvironment (TME), drivers of the immune response and mechanisms of tumor resistance to immunity. The development of novel immunotherapeutics and their use in combination with checkpoint inhibitors and other standard of care and novel treatment modalities is an area of particular attention across several tumor types, including melanoma, lung, ovarian, breast, pancreatic, renal, head and neck, brain and non-melanoma skin cancers. The 4th Immunotherapy Bridge meeting (28-29 November, 2018, Naples, Italy) focused on a wide range of evolving topics and trends in the field of cancer immunotherapy and key presentations from this meeting are summarised in this report.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Medical Oncology and Innovative Therapy, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Via Mariano Semmola, 80131, Naples, Italy.
| | - Carlo Bifulco
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Portland Medical Center, Portland, OR, USA
| | - Luigi Buonaguro
- Cancer Immunoregulation Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Leisha A Emens
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert L Ferris
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bernard A Fox
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Center in the Earle A. Chiles Research Institute at Providence Cancer Institute, Portland, Oregon, USA
| | - Greg M Delgoffe
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jérôme Galon
- National Institute of Health and Medical Research, INSERM, Cordeliers Research Center, Paris, France
| | - Cesare Gridelli
- Unit of Medical Oncology, Hospital "San Giuseppe Moscati", Avellino, Italy
| | - Marco Merlano
- Oncology Department, ASO Santa Croce e Carle Cuneo, Cuneo, Italy
| | - Paul Nathan
- Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| | - Kunle Odunsi
- Department of Gynaecologic Oncology, Executive Director, Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California San Francisco, Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, USA
| | - Sandro Pignata
- Uro-Gynaecological Department, Istituto Nazionale Tumori Fondazione G. Pascale, IRCCS, Naples, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Translational Immuno-oncology Laboratory, Yale Cancer Center, Medical Oncology, Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Stefani Spranger
- The Koch Institute for Integrative Cancer Research at MIT and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Gemelli, IRCCS, Rome, Italy
| | - Hassane Zarour
- Melanoma Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy Research Center, UCSF, San Francisco, California, USA.
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
536
|
Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Henschel V, Molinero L, Chui SY, Maiya V, Husain A, Winer EP, Loi S, Emens LA. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2019; 21:44-59. [PMID: 31786121 DOI: 10.1016/s1470-2045(19)30689-8] [Citation(s) in RCA: 825] [Impact Index Per Article: 137.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Immunotherapy in combination with chemotherapy has shown promising efficacy across many different tumour types. We report the prespecified second interim overall survival analysis of the phase 3 IMpassion130 study assessing the efficacy and safety of atezolizumab plus nab-paclitaxel in patients with unresectable, locally advanced or metastatic triple-negative breast cancer. METHODS In this randomised, placebo-controlled, double-blind, phase 3 trial, done in 246 academic centres and community oncology practices in 41 countries, patients aged 18 years or older, with previously untreated, histologically documented, locally advanced or metastatic triple-negative breast cancer, and Eastern Cooperative Oncology Group performance status of 0 or 1 were eligible. Patients were randomly assigned (1:1) using a permuted block method (block size of four) and an interactive voice-web response system. Randomisation was stratified by previous taxane use, liver metastases, and PD-L1 expression on tumour-infiltrating immune cells. Patients received atezolizumab 840 mg or matching placebo intravenously on day 1 and day 15 of every 28-day cycle and nab-paclitaxel 100 mg/m2 of body surface area intravenously on days 1, 8, and 15 until progression or unacceptable toxicity. Investigators, patients, and the funder were masked to treatment assignment. Coprimary endpoints were investigator-assessed progression-free survival per Response Evaluation Criteria in Solid Tumors version 1.1 and overall survival, assessed in the intention-to-treat population and in patients with PD-L1 immune cell-positive tumours (tumours with ≥1% PD-L1 expression). The final progression-free survival results were previously reported at the first interim overall survival analysis. The prespecified statistical testing hierarchy meant that overall survival in the subgroup of PD-L1 immune cell-positive patients could only be formally tested if overall survival was significantly different between the treatment groups in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT02425891. FINDINGS Between June 23, 2015, and May 24, 2017, 902 patients were enrolled, of whom 451 were randomly assigned to receive atezolizumab plus nab-paclitaxel and 451 were assigned to receive placebo plus nab-paclitaxel (the intention-to-treat population). Six patients from each group did not receive treatment. At the second interim analysis (data cutoff Jan 2, 2019), median follow-up was 18·5 months (IQR 9·6-22·8) in the atezolizumab group and 17·5 months (8·4-22·4) in the placebo group. Median overall survival in the intention-to-treat patients was 21·0 months (95% CI 19·0-22·6) with atezolizumab and 18·7 months (16·9-20·3) with placebo (stratified hazard ratio [HR] 0·86, 95% CI 0·72-1·02, p=0·078). In the exploratory overall survival analysis in patients with PD-L1 immune cell-positive tumours, median overall survival was 25·0 months (95% CI 19·6-30·7) with atezolizumab versus 18·0 months (13·6-20·1) with placebo (stratified HR 0·71, 0·54-0·94]). As of Sept 3, 2018 (the date up to which updated safety data were available), the most common grade 3-4 adverse events were neutropenia (38 [8%] of 453 patients in the atezolizumab group vs 36 [8%] of 437 patients in the placebo group), peripheral neuropathy (25 [6%] vs 12 [3%]), decreased neutrophil count (22 [5%] vs 16 [4%]), and fatigue (17 [4%] vs 15 [3%]). Treatment-related deaths occurred in two (<1%) patients in the atezolizumab group (autoimmune hepatitis related to atezolizumab [n=1] and septic shock related to nab-paclitaxel [n=1]) and one (<1%) patient in the placebo group (hepatic failure). No new treatment-related deaths have been reported since the primary clinical data cutoff date (April 17, 2018). INTERPRETATION Consistent with the first interim analysis, this second interim overall survival analysis of IMpassion130 indicates no significant difference in overall survival between the treatment groups in the intention-to-treat population but suggests a clinically meaningful overall survival benefit with atezolizumab plus nab-paclitaxel in patients with PD-L1 immune cell-positive disease. However, this positive result could not be formally tested due to the prespecified statistical testing hierarchy. For patients with PD-L1 immune cell-positive metastatic triple-negative breast cancer, atezolizumab plus nab-paclitaxel is an important therapeutic option in a disease with high unmet need. FUNDING F Hoffmann-La Roche and Genentech.
Collapse
Affiliation(s)
- Peter Schmid
- Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Sylvia Adams
- Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Carlos H Barrios
- Centro de Pesquisa Clínica, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil; Latin American Cooperative Oncology Group, Porto Alegre, Brazil; Grupo Oncoclínicas, Porto Alegre, Brazil
| | | | - Véronique Diéras
- Department of Medical Oncology, Institut Curie, Paris, France; Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | | | | | | | | | | | | | - Sherene Loi
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Leisha A Emens
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | | |
Collapse
|
537
|
Woodward WA. Building momentum for subsets of patients with advanced triple-negative breast cancer. Lancet Oncol 2019; 21:3-5. [PMID: 31786122 DOI: 10.1016/s1470-2045(19)30737-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Wendy A Woodward
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
538
|
Clinical Implications of Extracellular HMGA1 in Breast Cancer. Int J Mol Sci 2019; 20:ijms20235950. [PMID: 31779212 PMCID: PMC6928815 DOI: 10.3390/ijms20235950] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
The unconventional secretion of proteins is generally caused by cellular stress. During the tumorigenesis, tumor cells experience high levels of stress, and the secretion of some theoretically intracellular proteins is activated. Once in the extracellular space, these proteins play different paracrine and autocrine roles and could represent a vulnerability of cancer. One of these proteins is the high mobility group A1 (HMGA1), which is frequently overexpressed in tumors and presents a low expression in normal adult tissues. We have recently described that HMGA1 establishes an autocrine loop in invasive triple-negative breast cancer (TNBC) cells. The secretion of HMGA1 and its binding to the receptor for advanced glycation end products (RAGE) mediates the migration, invasion, and metastasis of TNBC cells and predicts the onset of metastasis in these patients. In this review, we summarized different strategies to exploit the novel tumorigenic phenotype mediated by extracellular HMGA1. We envisioned future clinical applications where the association between its change in subcellular localization and breast cancer progression could be used to predict tumor aggressiveness and guide treatment decisions. Furthermore, we proposed that targeting extracellular HMGA1 as monotherapy using monoclonal antibodies, or in combination with chemotherapy and other targeted therapies, could bring new therapeutic options for TNBC patients.
Collapse
|
539
|
Kim I, Sanchez K, McArthur HL, Page D. Immunotherapy in Triple-Negative Breast Cancer: Present and Future. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00345-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Abstract
Purpose of Review
Immunotherapy is emerging as an effective treatment option for metastatic triple-negative breast cancer. In this review, we summarize clinical data of immunotherapy in triple-negative breast cancer and comment on future directions in the field.
Recent Findings
IMpassion130 was a phase III trial that demonstrated progression-free survival benefit, and potentially overall survival benefit, of first-line chemotherapy (nab-paclitaxel) plus anti-programmed death ligand 1 (PD-L1) atezolizumab, among PD-L1-positive metastatic triple-negative breast cancers. Studies are ongoing to evaluate other combination therapies with immune checkpoint blockade in TNBC, and to evaluate efficacy in PD-L1-negative tumors and in later lines of therapy.
Summary
Immunotherapy is now a standard option in the treatment of triple-negative breast cancer. Ongoing trials may expand the degree of clinical benefit. Further work is ongoing to identify novel predictive biomarkers, which in the future may enable a personalized approach of combination immunotherapy.
Collapse
|
540
|
Lee JS, Yost SE, Blanchard S, Schmolze D, Yin HH, Pillai R, Robinson K, Tang A, Martinez N, Portnow J, Wen W, Yim JH, Brauer HA, Ren Y, Luu T, Mortimer J, Yuan Y. Phase I clinical trial of the combination of eribulin and everolimus in patients with metastatic triple-negative breast cancer. Breast Cancer Res 2019; 21:119. [PMID: 31703728 PMCID: PMC6839083 DOI: 10.1186/s13058-019-1202-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Alteration of the PI3K/AKT/mTOR pathway is a common genomic abnormality detected in triple-negative breast cancer (TNBC). Everolimus acts synergistically with eribulin in TNBC cell lines and xenograft models. This phase I trial was designed to test the safety and tolerability of combining eribulin and everolimus in patients with metastatic TNBC. METHODS The primary objective of this study was to evaluate the safety and toxicities of the combination. Patients with metastatic TNBC who had up to four lines of prior chemotherapies were enrolled. The combination of eribulin and everolimus was tested using three dosing levels: A1 (everolimus 5 mg daily; eribulin 1.4 mg/m2 days 1 and 8 every 3 weeks), A2 (everolimus 7.5 mg daily; eribulin 1.4 mg/m2, days 1 and 8 every 3 weeks), and B1 (everolimus 5 mg daily; eribulin 1.1 mg/m2 days 1 and 8 every 3 weeks). RESULTS Twenty-seven patients with median age 55 years were enrolled. Among 8 evaluable patients who received dose level A1, 4 had dose-limiting toxicities (DLTs). Among 3 evaluable patients treated with dose level A2, 2 had DLTs. Among 12 evaluable patients who received dose level B1, 4 had DLTs. The DLTs were neutropenia, stomatitis, and hyperglycemia. Over the study period, 59% had a ≥ grade 3 toxicity, 44% had ≥ grade 3 hematologic toxicities, and 22% had grade 4 hematologic toxicities. The most common hematological toxicities were neutropenia, leukopenia, and lymphopenia. Thirty-three percent had grade 3 non-hematologic toxicities. The most common non-hematological toxicities were stomatitis, hyperglycemia, and fatigue. The median number of cycles completed was 4 (range 0-8). Among 25 eligible patients, 9 patients (36%) achieved the best response as partial response, 9 (36%) had stable disease, and 7 (28%) had progression. The median time to progression was 2.6 months (95% CI [2.1, 4.0]), and median overall survival (OS) was 8.3 months (95% CI [5.5, undefined]). CONCLUSION Eribulin 1.1 mg/m2 days 1 and 8 every 3 weeks with everolimus 5 mg daily was defined as the highest dose with acceptable toxicity (RP2D). The combination is safe, and efficacy is modest. A post hoc analysis showed that participants that used dexamethasone mouthwash stayed on treatment for one additional cycle. TRIAL REGISTRATION ClinicalTrials.gov, NCT02120469. Registered 18 April 2014.
Collapse
Affiliation(s)
- Jin Sun Lee
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Susan E Yost
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Suzette Blanchard
- 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
| | - Hongwei Holly Yin
- Department of Pathology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Raju Pillai
- Department of Pathology, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Kim Robinson
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Aileen Tang
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Norma Martinez
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Jana Portnow
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Wei Wen
- Department of Surgery, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - John H Yim
- Department of Surgery, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | | | - Yuqi Ren
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Joanne Mortimer
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| | - Yuan Yuan
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center and Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
541
|
Yang J. Lack of Robust Prognostic Biomarkers for Immunotherapy in Breast Cancer-Adverse Events. JAMA Oncol 2019; 5:1639-1640. [PMID: 31513256 DOI: 10.1001/jamaoncol.2019.3596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Jiqiao Yang
- Department of Breast Surgery, Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
542
|
Gatti-Mays ME, Balko JM, Gameiro SR, Bear HD, Prabhakaran S, Fukui J, Disis ML, Nanda R, Gulley JL, Kalinsky K, Abdul Sater H, Sparano JA, Cescon D, Page DB, McArthur H, Adams S, Mittendorf EA. If we build it they will come: targeting the immune response to breast cancer. NPJ Breast Cancer 2019; 5:37. [PMID: 31700993 PMCID: PMC6820540 DOI: 10.1038/s41523-019-0133-7] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Historically, breast cancer tumors have been considered immunologically quiescent, with the majority of tumors demonstrating low lymphocyte infiltration, low mutational burden, and modest objective response rates to anti-PD-1/PD-L1 monotherapy. Tumor and immunologic profiling has shed light on potential mechanisms of immune evasion in breast cancer, as well as unique aspects of the tumor microenvironment (TME). These include elements associated with antigen processing and presentation as well as immunosuppressive elements, which may be targeted therapeutically. Examples of such therapeutic strategies include efforts to (1) expand effector T-cells, natural killer (NK) cells and immunostimulatory dendritic cells (DCs), (2) improve antigen presentation, and (3) decrease inhibitory cytokines, tumor-associated M2 macrophages, regulatory T- and B-cells and myeloid derived suppressor cells (MDSCs). The goal of these approaches is to alter the TME, thereby making breast tumors more responsive to immunotherapy. In this review, we summarize key developments in our understanding of antitumor immunity in breast cancer, as well as emerging therapeutic modalities that may leverage that understanding to overcome immunologic resistance.
Collapse
Affiliation(s)
- Margaret E. Gatti-Mays
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Justin M. Balko
- Department of Medicine and Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sofia R. Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Harry D. Bear
- Division of Surgical Oncology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA USA
| | - Sangeetha Prabhakaran
- Division of Surgical Oncology, Department of Surgery, University of New Mexico; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM USA
| | - Jami Fukui
- University of Hawaii Cancer Center, Honolulu, HI USA
| | | | - Rita Nanda
- The University of Chicago, Chicago, IL USA
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Kevin Kalinsky
- Columbia University Irving Medical Center, New York, NY USA
| | - Houssein Abdul Sater
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Joseph A. Sparano
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY USA
| | - David Cescon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON Canada
| | - David B. Page
- Providence Cancer Institute, Earle A. Chiles Research Institute, Portland, OR USA
| | | | - Sylvia Adams
- Perlmutter Cancer Center, NYU School of Medicine, New York, NY USA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, MA USA
- Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA USA
| |
Collapse
|
543
|
Molinero L, Li Y, Chang CW, Maund S, Berg M, Harrison J, Fassò M, O'Hear C, Hegde P, Emens LA. Tumor immune microenvironment and genomic evolution in a patient with metastatic triple negative breast cancer and a complete response to atezolizumab. J Immunother Cancer 2019; 7:274. [PMID: 31647026 PMCID: PMC6813065 DOI: 10.1186/s40425-019-0740-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/12/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metastatic TNBC (mTNBC) has a poor prognosis and few treatment options. The anti-PD-L1 antibody atezolizumab demonstrated clinical activity in mTNBC patients with PD-L1-positive tumor-infiltrating immune cells. The current study describes the tumor immune microenvironment (TiME) and genomic evolution across sequential therapies in a patient with a 31-year history of TNBC and a complete response (CR) to atezolizumab monotherapy. MATERIALS AND METHODS In 1986, the patient had surgery and radiotherapy (XRT) for newly diagnosed TNBC, followed by surgery and adjuvant chemotherapy for two locoregional recurrences. She developed mTNBC in 2009 and was sequentially treated with capecitabine, gemcitabine-carboplatin-iniparib (GCI), XRT and an experimental vaccine. She experienced disease progression (PD) to all these therapies. In 2013, she had a PD-L1 positive tumor and enrolled in a phase 1 atezolizumab monotherapy study (PCD4989g; NCT01375842). She received atezolizumab for 1 year with initial pseudo-progression followed by a partial response. After 1 year without treatment she experienced PD, reinitiated atezolizumab and subsequently achieved CR. Tumor specimens were collected at numerous times between 2008 and 2015 and assessed by immunohistochemistry, RNA-seq and DNA-seq. RESULTS TiME biomarkers, including CD8, ICs and PD-L1 on IC, increased after capecitabine and remained high after GCI, XRT and through pseudo-progression on atezolizumab. At PD post-atezolizumab exposure, TiME biomarkers decreased but PD-L1 status remained positive. Immune-related RNA signatures confirmed these findings. TNBC subtyping revealed evolution from luminal androgen receptor (LAR) to basal-like immune activated (BLIA). Genomic profiling showed truncal alterations in RB1 and TP53, while the presence of other genomic alterations varied over time. Tumor mutational burden peaked after XRT and declined after atezolizumab exposure. CONCLUSIONS This case report describes the evolution of TiME and TNBC molecular subtypes/genomics over time with sequential therapies in a TNBC patient with a CR to atezolizumab monotherapy. These data suggest the TiME is pliable and may be manipulated to maximize response to immunotherapy (NCT01375842, https://clinicaltrials.gov/ct2/show/NCT01375842?term=NCT01375842&rank=1 ).
Collapse
Affiliation(s)
- Luciana Molinero
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA.
| | - Yijin Li
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA
| | - Ching-Wei Chang
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA
| | - Sophia Maund
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA
| | - Maureen Berg
- School of Medicine, Oncology/Immunology, Johns Hopkins University, The Skip Viragh Outpatient Cancer Building, Floor 8, Viragh 8200-30, Box 11, 201 N Broadway, Baltimore, MD, 21287, USA
| | - Jeanne Harrison
- School of Medicine, Oncology/Immunology, Johns Hopkins University, The Skip Viragh Outpatient Cancer Building, Floor 8, Viragh 8200-30, Box 11, 201 N Broadway, Baltimore, MD, 21287, USA
| | - Marcella Fassò
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA
| | - Carol O'Hear
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA
| | - Priti Hegde
- Oncology Biomarker Development, PDCO-Immunolotherapy, Genentech, Incorporated, 1 DNA Way MS: 245c, South San Francisco, CA, 94080, USA
| | - Leisha A Emens
- School of Medicine, Oncology/Immunology, Johns Hopkins University, The Skip Viragh Outpatient Cancer Building, Floor 8, Viragh 8200-30, Box 11, 201 N Broadway, Baltimore, MD, 21287, USA. .,Bloomberg~Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, USA. .,University of Pittsburgh Medical Center, Hillman Cancer Center, 5117 Centre Avenue, Room 1.46e, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
544
|
Frank GA, Kuznetsova OA, Zavalishina LE, Andreeva YY, Moskvina LV. [Study of the PD-L1 status in breast cancer, by using the SP142 monoclonal antibody, and the prospects for determining treatment policy]. Arkh Patol 2019; 81:5-10. [PMID: 31626199 DOI: 10.17116/patol2019810515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate the expression of programmed death ligand 1 (PD-L1) in triple-negative and luminal B, HER2-negative breast cancer, by using the SP142 antibody and to assess the association of the PD-L1 status with prognosis for patients. MATERIAL AND METHODS The study was conducted using surgical materials (full sections) obtained from 72 patients. The sections were stained with the SP142 PD-L1 antibody. RESULTS Differences were found in the detection rates of the PD-L1-positive status in the primary tumor and regional metastasis (primary tumor in 26 (36.1%) cases and metastasis in 18 (47.4%) of the 38 cases). By and large, the PD-L1-positive status was less common in the patients receiving neoadjuvant chemotherapy (25.8% versus 53.7%); however, it should be noted that the PD-L1-positive status was more often detected in those who had a higher residual cancer burden (RCB) (RCB-III versus RCB-II). CONCLUSION Considering the findings, it is necessary to clarify the status of not only a primary focus, but also clinically significant metastases, especially if the primary tumor has yielded a negative result. The patients with disease progression who receive standard therapy regimens may have a chance for a good result when using PD-1/PD-L1 blockers. At the same time, the association of the PD-L1 status with RCB may affect the choice of adjuvant treatment policy.
Collapse
Affiliation(s)
- G A Frank
- Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia, Moscow, Russia
| | - O A Kuznetsova
- Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia, Moscow, Russia
| | - L E Zavalishina
- Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia, Moscow, Russia
| | - Yu Yu Andreeva
- Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia, Moscow, Russia
| | - L V Moskvina
- Russian Medical Academy of Continuing Professional Education, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
545
|
Page DB, Bear H, Prabhakaran S, Gatti-Mays ME, Thomas A, Cobain E, McArthur H, Balko JM, Gameiro SR, Nanda R, Gulley JL, Kalinsky K, White J, Litton J, Chmura SJ, Polley MY, Vincent B, Cescon DW, Disis ML, Sparano JA, Mittendorf EA, Adams S. Two may be better than one: PD-1/PD-L1 blockade combination approaches in metastatic breast cancer. NPJ Breast Cancer 2019; 5:34. [PMID: 31602395 PMCID: PMC6783471 DOI: 10.1038/s41523-019-0130-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/05/2019] [Indexed: 01/07/2023] Open
Abstract
Antibodies blocking programmed death 1 (anti-PD-1) or its ligand (anti-PD-L1) are associated with modest response rates as monotherapy in metastatic breast cancer, but are generally well tolerated and capable of generating dramatic and durable benefit in a minority of patients. Anti-PD-1/L1 antibodies are also safe when administered in combination with a variety of systemic therapies (chemotherapy, targeted therapies), as well as with radiotherapy. We summarize preclinical, translational, and preliminary clinical data in support of combination approaches with anti-PD-1/L1 in metastatic breast cancer, focusing on potential mechanisms of synergy, and considerations for clinical practice and future investigation.
Collapse
Affiliation(s)
- David B. Page
- Providence Cancer Institute; Earle A. Chiles Research Institute, Portland, OR USA
| | - Harry Bear
- Division of Surgical Oncology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA USA
| | - Sangeetha Prabhakaran
- Department of Surgery, Division of Surgery, University of New Mexico; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM USA
| | | | - Alexandra Thomas
- Wake Forest University School of Medicine, Winston-Salem, NC USA
| | | | | | - Justin M. Balko
- Department of Medicine and Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sofia R. Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, MD USA
| | - Rita Nanda
- The University of Chicago, Chicago, IL USA
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | | | - Julia White
- Ohio State Wexner Medical Center, Columbus, OH USA
| | | | | | | | | | - David W. Cescon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON Canada
| | | | - Joseph A. Sparano
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY USA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital; Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA USA
| | - Sylvia Adams
- Perlmutter Cancer Center, NYU School of Medicine, New York, NY USA
| |
Collapse
|
546
|
Targeting PD-1 in cancer: Biological insights with a focus on breast cancer. Crit Rev Oncol Hematol 2019; 142:35-43. [DOI: 10.1016/j.critrevonc.2019.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/09/2019] [Accepted: 07/14/2019] [Indexed: 12/25/2022] Open
|
547
|
Márquez-Garbán DC, Deng G, Comin-Anduix B, Garcia AJ, Xing Y, Chen HW, Cheung-Lau G, Hamilton N, Jung ME, Pietras RJ. Antiestrogens in combination with immune checkpoint inhibitors in breast cancer immunotherapy. J Steroid Biochem Mol Biol 2019; 193:105415. [PMID: 31226312 PMCID: PMC6903431 DOI: 10.1016/j.jsbmb.2019.105415] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/06/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022]
Abstract
Breast cancers (BCs) with expression of estrogen receptor-alpha (ERα) occur in more than 70% of newly-diagnosed patients in the U.S. Endocrine therapy with antiestrogens or aromatase inhibitors is an important intervention for BCs that express ERα, and it remains one of the most effective targeted treatment strategies. However, a substantial proportion of patients with localized disease, and essentially all patients with metastatic BC, become resistant to current endocrine therapies. ERα is present in most resistant BCs, and in many of these its activity continues to regulate BC growth. Fulvestrant represents one class of ERα antagonists termed selective ER downregulators (SERDs). Treatment with fulvestrant causes ERα down-regulation, an event that helps overcome several resistance mechanisms. Unfortunately, full antitumor efficacy of fulvestrant is limited by its poor bioavailability in clinic. We have designed and tested a new generation of steroid-like SERDs. Using ERα-positive BC cells in vitro, we find that these compounds suppress ERα protein levels with efficacy similar to fulvestrant. Moreover, these new SERDs markedly inhibit ERα-positive BC cell transcription and proliferation in vitro even in the presence of estradiol-17β. In vivo, the SERD termed JD128 significantly inhibited tumor growth in MCF-7 xenograft models in a dose-dependent manner (P < 0.001). Further, our findings indicate that these SERDs also interact with ER-positive immune cells in the tumor microenvironment such as myeloid-derived suppressor cells (MDSC), tumor infiltrating lymphocytes and other selected immune cell subpopulations. SERD-induced inhibition of MDSCs and concurrent actions on CD8+ and CD4 + T-cells promotes interaction of immune checkpoint inhibitors with BC cells in preclinical models, thereby leading to enhanced tumor killing even among highly aggressive BCs such as triple-negative BC that lack ERα expression. Since monotherapy with immune checkpoint inhibitors has not been effective for most BCs, combination therapies with SERDs that enhance immune recognition may increase immunotherapy responses in BC and improve patient survival. Hence, ERα antagonists that also promote ER downregulation may potentially benefit patients who are unresponsive to current endocrine therapies.
Collapse
Affiliation(s)
- Diana C Márquez-Garbán
- UCLA David Geffen School of Medicine, Department of Medicine, Division of Hematology-Oncology, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Gang Deng
- UCLA Department of Chemistry and Biochemistry, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Begonya Comin-Anduix
- UCLA Department of Surgery, Division of Surgical Oncology, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Alejandro J Garcia
- UCLA David Geffen School of Medicine, Department of Medicine, Division of Hematology-Oncology, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Yanpeng Xing
- UCLA Department of Chemistry and Biochemistry, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Hsiao-Wang Chen
- UCLA David Geffen School of Medicine, Department of Medicine, Division of Hematology-Oncology, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Gardenia Cheung-Lau
- UCLA Department of Surgery, Division of Surgical Oncology, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Nalo Hamilton
- UCLA School of Nursing, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Michael E Jung
- UCLA Department of Chemistry and Biochemistry, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA
| | - Richard J Pietras
- UCLA David Geffen School of Medicine, Department of Medicine, Division of Hematology-Oncology, Los Angeles CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, Los Angeles CA 90095, USA.
| |
Collapse
|
548
|
Passariello M, D'Alise AM, Esposito A, Vetrei C, Froechlich G, Scarselli E, Nicosia A, De Lorenzo C. Novel Human Anti-PD-L1 mAbs Inhibit Immune-Independent Tumor Cell Growth and PD-L1 Associated Intracellular Signalling. Sci Rep 2019; 9:13125. [PMID: 31511565 PMCID: PMC6739323 DOI: 10.1038/s41598-019-49485-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023] Open
Abstract
The novel antibody-based immunotherapy in oncology exploits the activation of immune system mediated by immunomodulatory antibodies specific for immune checkpoints. Among them, the programmed death ligand-1 (PD-L1) is of particular interest as it is expressed not only on T-cells, but also on other immune cells and on a large variety of cancer cells, such as breast cancer cells, considering its high expression in both ErbB2-positive and Triple Negative Breast Cancers. We demonstrate here that PD-L1_1, a novel anti-PD-L1 T -cell stimulating antibody, inhibits PD-L1-tumor cell growth also by affecting the intracellular MAPK pathway and by activating caspase 3. Similar in vitro results were obtained for the first time here also with the clinically validated anti-PD-L1 mAb Atezolizumab and in vivo with another validated anti-mouse anti-PD-L1 mAb. Moreover, we found that two high affinity variants of PD-L1_1 inhibited tumor cell viability more efficiently than the parental PD-L1_1 by affecting the same MAPK pathways with a more potent effect. Altogether, these results shed light on the role of PD-L1 in cancer cells and suggest that PD-L1_1 and its high affinity variants could become powerful antitumor weapons to be used alone or in combination with other drugs such as the anti-ErbB2 cAb already successfully tested in in vitro combinatorial treatments.
Collapse
Affiliation(s)
- Margherita Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | | | - Annachiara Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Cinzia Vetrei
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Guendalina Froechlich
- Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.,European School of Molecular Medicine, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | | | - Alfredo Nicosia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.,Keires AG Bäumleingasse 18, CH-4051, Basel, Switzerland
| | - Claudia De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy. .,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.
| |
Collapse
|
549
|
Sceneay J, Goreczny GJ, Wilson K, Morrow S, DeCristo MJ, Ubellacker JM, Qin Y, Laszewski T, Stover DG, Barrera V, Hutchinson JN, Freedman RA, Mittendorf EA, McAllister SS. Interferon Signaling Is Diminished with Age and Is Associated with Immune Checkpoint Blockade Efficacy in Triple-Negative Breast Cancer. Cancer Discov 2019; 9:1208-1227. [PMID: 31217296 PMCID: PMC11167954 DOI: 10.1158/2159-8290.cd-18-1454] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/16/2019] [Accepted: 06/14/2019] [Indexed: 11/16/2022]
Abstract
Immune checkpoint blockade (ICB) therapy, which targets T cell-inhibitory receptors, has revolutionized cancer treatment. Among the breast cancer subtypes, evaluation of ICB has been of greatest interest in triple-negative breast cancer (TNBC) due to its immunogenicity, as evidenced by the presence of tumor-infiltrating lymphocytes and elevated PD-L1 expression relative to other subtypes. TNBC incidence is equally distributed across the age spectrum, affecting 10% to 15% of women in all age groups. Here we report that increased immune dysfunction with age limits ICB efficacy in aged TNBC-bearing mice. The tumor microenvironment in both aged mice and patients with TNBC shows decreased IFN signaling and antigen presentation, suggesting failed innate immune activation with age. Triggering innate immune priming with a STING agonist restored response to ICB in aged mice. Our data implicate age-related immune dysfunction as a mechanism of ICB resistance in mice and suggest potential prognostic utility of assessing IFN-related genes in patients with TNBC receiving ICB therapy. SIGNIFICANCE: These data demonstrate for the first time that age determines the T cell-inflamed phenotype in TNBC and affects response to ICB in mice. Evaluating IFN-related genes from tumor genomic data may aid identification of patients for whom combination therapy including an IFN pathway activator with ICB may be required.This article is highlighted in the In This Issue feature, p. 1143.
Collapse
Affiliation(s)
- Jaclyn Sceneay
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Gregory J Goreczny
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kristin Wilson
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sara Morrow
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Molly J DeCristo
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jessalyn M Ubellacker
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Yuanbo Qin
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Tyler Laszewski
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Daniel G Stover
- Division of Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Victor Barrera
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - John N Hutchinson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rachel A Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sandra S McAllister
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
550
|
Tabrizi S, McDuff S, Ho AY. Combining Radiation Therapy with Immune Checkpoint Blockadein Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00327-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|