251
|
Adaptive immune resistance at the tumour site: mechanisms and therapeutic opportunities. Nat Rev Drug Discov 2022; 21:529-540. [PMID: 35701637 DOI: 10.1038/s41573-022-00493-5] [Citation(s) in RCA: 207] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2022] [Indexed: 12/11/2022]
Abstract
Tumours employ various tactics to adapt and eventually resist immune attack. These mechanisms are collectively called adaptive immune resistance (AIR). The first defined and therapeutically validated AIR mechanism is the selective induction of programmed cell death 1 ligand 1 (PDL1) by interferon-γ in the tumour. Blockade of PDL1 binding to its receptor PD1 by antibodies (anti-PD therapy) has resulted in remission of a fraction of patients with advanced-stage cancer, especially in solid tumours. However, many clinical trials combining anti-PD therapy with other antitumour drugs conducted without a strong mechanistic rationale have failed to identify a synergistic or additive effect. In this Perspective article, we discuss why defining AIR mechanisms at the tumour site should be a key focus to direct future drug development as well as practical approaches to improve current cancer therapy.
Collapse
|
252
|
Uchimiak K, Badowska-Kozakiewicz AM, Sobiborowicz-Sadowska A, Deptała A. Current State of Knowledge on the Immune Checkpoint Inhibitors in Triple-Negative Breast Cancer Treatment: Approaches, Efficacy, and Challenges. Clin Med Insights Oncol 2022; 16:11795549221099869. [PMID: 35721387 PMCID: PMC9201309 DOI: 10.1177/11795549221099869] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with limited treatment options. Recently, there has been a growing interest in immunotherapy with immune checkpoint inhibitors (ICIs) in TNBC, leading to extensive preclinical and clinical research. This review summarizes the current state of knowledge on ICIs efficacy and their predictive markers in TNBC and highlights the areas where the data are still limited. Currently, the only approved ICI-based regimen for TNBC is pembrolizumab with chemotherapy. Its advantage over chemotherapy alone was confirmed for non-metastatic TNBC regardless of programmed death-ligand 1 (PD-L1) expression (KEYNOTE-522) and for metastatic, PD-L1-positive TNBC (KEYNOTE-355). Pembrolizumab's efficacy was also evaluated in monotherapy, or in combination with niraparib and radiation therapy, showing potential efficacy and acceptable safety profile in phase 2 clinical trials. Atezolizumab + nab-paclitaxel increased the overall survival (OS) over placebo + nab-paclitaxel in early TNBC, regardless of PD-L1 status (IMpassion031). In IMpassion130 (untreated, advanced TNBC), the OS improvement was not statistically significant in the intention-to-treat population but clinically meaningful in the PD-L1 positive cohort. The durvalumab-anthracycline combination showed an increased response durability over placebo anthracycline in early TNBC (GeparNuevo). Several phase 1 clinical trials also showed a potential efficacy of atezolizumab and avelumab monotherapy in metastatic TNBC. ICIs appear to be applicable in both neoadjuvant and adjuvant settings, and are both pretreated and previously untreated patients. Further research is necessary to determine the most beneficial drug combinations and optimize patient selection. It is essential to identify the predictive markers for ICIs and factors affecting their expression.
Collapse
Affiliation(s)
- Katarzyna Uchimiak
- Students’ Scientific Organization of
Cancer Cell Biology, Department of Cancer Prevention, Medical University of Warsaw,
Warsaw, Poland
| | | | - Aleksandra Sobiborowicz-Sadowska
- Students’ Scientific Organization of
Cancer Cell Biology, Department of Cancer Prevention, Medical University of Warsaw,
Warsaw, Poland
| | - Andrzej Deptała
- Department of Cancer Prevention,
Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
253
|
Zambelli A, Sgarra R, De Sanctis R, Agostinetto E, Santoro A, Manfioletti G. Heterogeneity of triple-negative breast cancer: understanding the Daedalian labyrinth and how it could reveal new drug targets. Expert Opin Ther Targets 2022; 26:557-573. [PMID: 35638300 DOI: 10.1080/14728222.2022.2084380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is considered the most aggressive breast cancer subtype with the least favorable outcomes. However, recent research efforts have generated an enhanced knowledge of the biology of the disease and have provided a new, more comprehensive understanding of the multifaceted ecosystem that underpins TNBC. AREAS COVERED In this review, the authors illustrate the principal biological characteristics of TNBC, the molecular driver alterations, targetable genes, and the biomarkers of immune engagement that have been identified across the subgroups of TNBC. Accordingly, the authors summarize the landscape of the innovative and investigative biomarker-driven therapeutic options in TNBC that emerge from the unique biological basis of the disease. EXPERT OPINION The therapeutic setting of TNBC is rapidly evolving. An enriched understanding of the tumor spatial and temporal heterogeneity and the surrounding microenvironment of this complex disease can effectively support the development of novel and tailored opportunities of treatment.
Collapse
Affiliation(s)
- Alberto Zambelli
- Medical Oncology and Hematology Unit, IRCCS - Humanitas Clinical and Research Center, Humanitas Cancer Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Riccardo Sgarra
- Department of Life sciences, University of Trieste, Trieste, Italy
| | - Rita De Sanctis
- Medical Oncology and Hematology Unit, IRCCS - Humanitas Clinical and Research Center, Humanitas Cancer Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elisa Agostinetto
- Department of Biomedical Sciences, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium and Humanitas University, Milan, Italy
| | - Armando Santoro
- Medical Oncology and Hematology Unit, IRCCS - Humanitas Clinical and Research Center, Humanitas Cancer Center, Milan, Italy
| | | |
Collapse
|
254
|
Čeprnja T, Mrklić I, Perić Balja M, Marušić Z, Blažićević V, Spagnoli GC, Juretić A, Čapkun V, Tečić Vuger A, Vrdoljak E, Tomić S. Prognostic Significance of Lymphocyte Infiltrate Localization in Triple-Negative Breast Cancer. J Pers Med 2022; 12:941. [PMID: 35743725 PMCID: PMC9224650 DOI: 10.3390/jpm12060941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
High infiltration by tumor-infiltrating lymphocytes (TILs) is associated with favorable prognosis in different tumor types, but the clinical significance of their spatial localization within the tumor microenvironment is debated. To address this issue, we evaluated the accumulation of intratumoral TILs (itTILs) and stromal TILs (sTILs) in samples from 97 patients with early triple-negative breast cancer (TNBC) in the center (sTIL central) and periphery (sTIL peripheral) of tumor tissues. Moreover, the presence of primary and secondary lymphoid aggregates (LAs) and the expression levels of the cancer testis antigen (CTA), NY-ESO-1, and PD-L1 were explored. High infiltration by itTILs was observed in 12/97 samples (12.3%), unrelated to age, Ki67 expression, tumor size, histologic type and grade, and LA presence. NY-ESO-1 was expressed in tumor cells in 37 samples (38%), with a trend suggesting a correlation with itTIL infiltration (p = 0.0531). PD-L1 expression was detected in immune cells in 47 samples (49%) and was correlated with histologic grade, sTILs, and LA formation. The presence of primary LAs was significantly correlated with better disease-free survival (DFS) (p = 0.027). Moreover, no tumor progression was observed during >40 months of clinical follow up in the 12 patients with high itTILs or in the 14 patients with secondary LAs. Thus, careful evaluation of lymphoid infiltrate intratumoral localization might provide important prognostic information.
Collapse
Affiliation(s)
- Toni Čeprnja
- Department of Pathology, Forensic Medicine and Cytology, University Hospital Center Split, School of Medicine, University of Split, 21000 Split, Croatia; (I.M.); (S.T.)
| | - Ivana Mrklić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital Center Split, School of Medicine, University of Split, 21000 Split, Croatia; (I.M.); (S.T.)
| | - Melita Perić Balja
- Department of Pathology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Zlatko Marušić
- Department of Pathology, Zagreb University Hospital Center, 10000 Zagreb, Croatia;
| | | | | | - Antonio Juretić
- Department of Oncology, Clinical Hospital “Sveti Duh”, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Vesna Čapkun
- Department of Nuclear Medicine, University Hospital Centre Split, School of Medicine, University of Split, 21000 Split, Croatia;
| | - Ana Tečić Vuger
- Department of Oncology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Eduard Vrdoljak
- Department of Oncology, University Hospital Center Split, University of Split, 21000 Split, Croatia;
| | - Snježana Tomić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital Center Split, School of Medicine, University of Split, 21000 Split, Croatia; (I.M.); (S.T.)
| |
Collapse
|
255
|
Ascierto PA, Avallone A, Bhardwaj N, Bifulco C, Bracarda S, Brody JD, Buonaguro L, Demaria S, Emens LA, Ferris RL, Galon J, Khleif SN, Klebanoff CA, Laskowski T, Melero I, Paulos CM, Pignata S, Ruella M, Svane IM, Taube JM, Fox BA, Hwu P, Puzanov I. Perspectives in Immunotherapy: meeting report from the Immunotherapy Bridge, December 1st-2nd, 2021. J Transl Med 2022; 20:257. [PMID: 35672823 PMCID: PMC9172186 DOI: 10.1186/s12967-022-03471-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/30/2022] [Indexed: 01/16/2023] Open
Abstract
Over the past decade, immunotherapy has become an increasingly fundamental modality in the treatment of cancer. The positive impact of immune checkpoint inhibition, especially anti-programmed death (PD)-1/PD-ligand (L)1 blockade, in patients with different cancers has focused attention on the potential for other immunotherapeutic approaches. These include inhibitors of additional immune checkpoints, adoptive cell transfer (ACT), and therapeutic vaccines. Patients with advanced cancers who previously had limited treatment options available may now benefit from immunotherapies that can offer durable responses and improved survival outcomes. However, despite this, a significant proportion of patients fail to respond to immunotherapy, especially those with less immunoresponsive cancer types, and there remains a need for new treatment strategies.The virtual Immunotherapy Bridge (December 1st-2nd, 2021), organized by the Fondazione Melanoma Onlus, Naples, Italy in collaboration with the Society for Immunotherapy of Cancer addressed several areas of current research in immunotherapy, including lessons learned from cell therapies, drivers of immune response, and trends in immunotherapy across different cancers, and these are summarised here.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlo Bifulco
- Providence Genomics and Earle A. Chiles Research Institute, Portland, OR, USA
| | - Sergio Bracarda
- Medical and Translational Oncology Unit, Department of Oncology, Azienda Ospedaliera Santa Maria, Terni, Italy
| | - Joshua D Brody
- Department of Medicine, Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luigi Buonaguro
- Department of Experimental Oncology, Innovative Immunological Models Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College; Sandra and Edward Meyer Cancer Center; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Leisha A Emens
- Magee Women's Hospital/UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology/Equipe Labellisée Ligue Contre Le Cancer/Centre de Recherche Des Cordeliers, Sorbonne Université, Université Paris Cité, Marseille, France
| | - Samir N Khleif
- The Loop Immuno Oncology Laboratory, Georgetown University Medical School, Washington, DC, USA
| | - Christopher A Klebanoff
- Human Oncology and Pathogenesis Program, Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center (MSKCC)/Center for Cell Engineering, MSKCC/Parker Institute for Cancer Immunotherapy/Weill Cornell Medical College, New York, NY, USA
| | - Tamara Laskowski
- Head of New Therapeutic Products - Personalized Medicine, Lonza Global, Houston, TX, USA
| | - Ignacio Melero
- Department of Immunology and Immunotherapy, Clinica Universidad de Navarra and CIBERONC, Pamplona, Spain
| | | | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Marco Ruella
- Center for Cellular Immunotherapies and Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins University SOM, Baltimore, MD, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Cancer Institute, Portland, OR, USA
| | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
256
|
Rizzo A, Cusmai A, Massafra R, Bove S, Comes MC, Fanizzi A, Rinaldi L, Acquafredda S, Gadaleta-Caldarola G, Oreste D, Zito A, Giotta F, Lorusso V, Palmiotti G. Pathological Complete Response to Neoadjuvant Chemoimmunotherapy for Early Triple-Negative Breast Cancer: An Updated Meta-Analysis. Cells 2022; 11:1857. [PMID: 35740985 PMCID: PMC9221459 DOI: 10.3390/cells11121857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have made a breakthrough in the systemic treatment for metastatic triple-negative breast cancer (TNBC) patients. However, results of phase II and III clinical trials assessing ICIs plus chemotherapy as neoadjuvant treatment were controversial and conflicting. We performed a meta-analysis aimed at assessing the Odds Ratio (OR) of the pathological complete response (pCR) rate in trials assessing neoadjuvant chemoimmunotherapy in TNBC. According to our results, the use of neoadjuvant chemoimmunotherapy was associated with higher pCR (OR 1.95; 95% Confidence Intervals, 1.27-2.99). In addition, we highlighted that this benefit was observed regardless of PD-L1 status since the analysis reported a statistically significant and clinically meaningful benefit in both PD-L1 positive and PD-L1 negative patients. These findings further support the exploration of the role of ICIs plus chemotherapy in early-stage TNBC, given the potentially meaningful clinical impact of these agents. Further studies aimed at more deeply investigating this emerging topic in breast cancer immunotherapy are warranted.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (L.R.); (S.A.); (G.P.)
| | - Antonio Cusmai
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (L.R.); (S.A.); (G.P.)
| | - Raffaella Massafra
- Struttura Semplice Dipartimentale di Fisica Sanitaria, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (R.M.); (S.B.); (M.C.C.); (A.F.)
| | - Samantha Bove
- Struttura Semplice Dipartimentale di Fisica Sanitaria, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (R.M.); (S.B.); (M.C.C.); (A.F.)
| | - Maria Colomba Comes
- Struttura Semplice Dipartimentale di Fisica Sanitaria, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (R.M.); (S.B.); (M.C.C.); (A.F.)
| | - Annarita Fanizzi
- Struttura Semplice Dipartimentale di Fisica Sanitaria, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (R.M.); (S.B.); (M.C.C.); (A.F.)
| | - Lucia Rinaldi
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (L.R.); (S.A.); (G.P.)
| | - Silvana Acquafredda
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (L.R.); (S.A.); (G.P.)
| | - Gennaro Gadaleta-Caldarola
- Medical Oncology Unit, ‘Mons. R. Dimiccoli’ Hospital, Barletta (BT), Azienda Sanitaria Locale Barletta, 76121 Barletta, Italy;
| | - Donato Oreste
- Radiology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Giovanni Paolo II, 70124 Bari, Italy;
| | - Alfredo Zito
- Unità Operativa Complessa di Anatomia Patologica, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy;
| | - Francesco Giotta
- Unità Operativa Complessa di Oncologia Medica, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (F.G.); (V.L.)
| | - Vito Lorusso
- Unità Operativa Complessa di Oncologia Medica, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (F.G.); (V.L.)
| | - Gennaro Palmiotti
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, IRCCS, Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (L.R.); (S.A.); (G.P.)
| |
Collapse
|
257
|
Zacharakis N, Huq LM, Seitter SJ, Kim SP, Gartner JJ, Sindiri S, Hill VK, Li YF, Paria BC, Ray S, Gasmi B, Lee CC, Prickett TD, Parkhurst MR, Robbins PF, Langhan MM, Shelton TE, Parikh AY, Levi ST, Hernandez JM, Hoang CD, Sherry RM, Yang JC, Feldman SA, Goff SL, Rosenberg SA. Breast Cancers Are Immunogenic: Immunologic Analyses and a Phase II Pilot Clinical Trial Using Mutation-Reactive Autologous Lymphocytes. J Clin Oncol 2022; 40:1741-1754. [PMID: 35104158 PMCID: PMC9148699 DOI: 10.1200/jco.21.02170] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/08/2021] [Accepted: 12/20/2021] [Indexed: 07/30/2023] Open
Abstract
PURPOSE Metastatic breast cancer (mBrCa) is most often an incurable disease with only modest responses to available immunotherapies. This study investigates the immunogenicity of somatic mutations in breast cancer and explores the therapeutic efficacy in a pilot trial of mutation-reactive tumor-infiltrating lymphocytes (TILs) in patients with metastatic disease. PATIENTS AND METHODS Forty-two patients with mBrCa refractory to previous lines of treatment underwent surgical resection of a metastatic lesion(s), isolation of TIL cultures, identification of exomic nonsynonymous tumor mutations, and immunologic screening for neoantigen reactivity. Clinically eligible patients with appropriate reactivity were enrolled into one cohort of an ongoing phase II pilot trial of adoptive cell transfer of selected neoantigen-reactive TIL, with a short course of pembrolizumab (ClinicalTrials.gov identifier: NCT01174121). RESULTS TILs were isolated and grown in culture from the resected lesions of all 42 patients with mBrCa, and a median number of 112 (range: 6-563) nonsynonymous mutations per patient were identified. Twenty-eight of 42 (67%) patients contained TIL that recognized at least one immunogenic somatic mutation (median: 3 neoantigens per patient, range: 1-11), and 13 patients demonstrated robust reactivity appropriate for adoptive transfer. Eight patients remained clinically eligible for treatment, and six patients were enrolled on a protocol of adoptive cell transfer of enriched neoantigen-specific TIL, in combination with pembrolizumab (≤ 4 doses). Objective tumor regression was noted in three patients, including one complete response (now ongoing over 5.5 years) and two partial responses (6 and 10 months). CONCLUSION Most patients with breast cancer generated a natural immune response targeting the expressed products of their cancer mutations. Adoptive transfer of TIL is a highly personalized experimental option for patients with mBrCa shown to be capable of mediating objective responses in this pilot trial and deserves further study.
Collapse
Affiliation(s)
- Nikolaos Zacharakis
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lutfi M. Huq
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Samantha J. Seitter
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sanghyun P. Kim
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jared J. Gartner
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sivasish Sindiri
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Victoria K. Hill
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yong F. Li
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Biman C. Paria
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Satyajit Ray
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Billel Gasmi
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Chyi-chia Lee
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Todd D. Prickett
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maria R. Parkhurst
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Paul F. Robbins
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michelle M. Langhan
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Thomas E. Shelton
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Anup Y. Parikh
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Shoshana T. Levi
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jonathan M. Hernandez
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Chuong D. Hoang
- Thoracic Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Richard M. Sherry
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Lyell Immunopharma, South San Francisco, CA
| | - James C. Yang
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Steven A. Feldman
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA
| | - Stephanie L. Goff
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
258
|
Howard FM, Villamar D, He G, Pearson AT, Nanda R. The emerging role of immune checkpoint inhibitors for the treatment of breast cancer. Expert Opin Investig Drugs 2022; 31:531-548. [PMID: 34569400 PMCID: PMC8995399 DOI: 10.1080/13543784.2022.1986002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Breast cancer has traditionally been viewed as immunogenically 'cold,' but two immune checkpoint inhibitors have been approved in combination with chemotherapy for PD-L1 positive advanced triple-negative breast cancer (TNBC), and pembrolizumab was also recently approved for early stage TNBC. As the landscape is rapidly evolving, a comprehensive review of checkpoint inhibitors in breast cancer is needed to aid clinicians in selecting appropriate candidates for therapy, and to highlight ongoing promising studies in this area and topics in need of further investigation. AREA COVERED This review summarizes the latest evidence from completed and ongoing trials of immune checkpoint inhibitors. Ongoing studies were identified using a search of ClinicalTrials.gov with the term 'breast cancer' along with specific checkpoint inhibitor agents. EXPERT OPINION A number of novel combination strategies are under investigation to enhance response and overcome resistance to immunotherapy, with promising preliminary data from checkpoint inhibitors targeting TIGIT, combinations with small molecule inhibitors such as lenvatinib, and injectable agents directly influencing the immune microenvironment. As immunotherapy enters into the curative setting, biomarkers predictive of immunotherapy benefit are needed, as PD-L1 status has not been a helpful discriminator in completed trials in early-stage breast cancer.
Collapse
Affiliation(s)
| | - Dario Villamar
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Gong He
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Rita Nanda
- Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
259
|
Kothari G. Role of radiomics in predicting immunotherapy response. J Med Imaging Radiat Oncol 2022; 66:575-591. [PMID: 35581928 PMCID: PMC9323544 DOI: 10.1111/1754-9485.13426] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022]
Abstract
Immunotherapies have revolutionised cancer management. Despite their success, durable responses are limited to a subset of patients. Prediction of immunotherapy response in patients has proven to be difficult due to a lack of robust biomarkers. Routinely collected imaging may offer an additional information source to personalise patient treatment, with advantages over tissue-based biomarkers. Quantitative image analysis or radiomics, which involves the high-throughput extraction of imaging features, has the potential to non-invasively predict cancer histology, outcomes and prognosis. This review evaluates the value of radiomics in patients undergoing immunotherapy, with a summary provided of the performance of radiomics models in predicting immunotherapy response and toxicity, as well as immune correlates. Much of the literature focussed on clinical endpoints and correlates to tissue biomarkers, particularly in lung cancer, while few studies investigated association with immune-related adverse events. Strengths of the studies included more frequent use of clinical trial datasets, homogenous patient cohorts and high-quality diagnostic scans. Limitations of the studies include heterogeneity in study methodology, lack of well-defined homogenous imaging datasets, limited open publishing of imaging datasets, coding and parameters used for radiomics signature development and limited use of external validation datasets. Future research should address the above limitations, as well as further explore the relationship between radiomics and immune-related adverse effects and less well-studied biological correlates such tumour mutational burden, and incorporate known clinical prognostic scores into radiomics models.
Collapse
Affiliation(s)
- Gargi Kothari
- Department of Radiation OncologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of Oncology, University of MelbournePeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| |
Collapse
|
260
|
Haist M, Mailänder V, Bros M. Nanodrugs Targeting T Cells in Tumor Therapy. Front Immunol 2022; 13:912594. [PMID: 35693776 PMCID: PMC9174908 DOI: 10.3389/fimmu.2022.912594] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
In contrast to conventional anti-tumor agents, nano-carriers allow co-delivery of distinct drugs in a cell type-specific manner. So far, many nanodrug-based immunotherapeutic approaches aim to target and kill tumor cells directly or to address antigen presenting cells (APC) like dendritic cells (DC) in order to elicit tumor antigen-specific T cell responses. Regulatory T cells (Treg) constitute a major obstacle in tumor therapy by inducing a pro-tolerogenic state in APC and inhibiting T cell activation and T effector cell activity. This review aims to summarize nanodrug-based strategies that aim to address and reprogram Treg to overcome their immunomodulatory activity and to revert the exhaustive state of T effector cells. Further, we will also discuss nano-carrier-based approaches to introduce tumor antigen-specific chimeric antigen receptors (CAR) into T cells for CAR-T cell therapy which constitutes a complementary approach to DC-focused vaccination.
Collapse
Affiliation(s)
| | | | - Matthias Bros
- University Medical Center Mainz, Department of Dermatology, Mainz, Germany
| |
Collapse
|
261
|
Immune Checkpoint Blockades in Triple-Negative Breast Cancer: Current State and Molecular Mechanisms of Resistance. Biomedicines 2022; 10:biomedicines10051130. [PMID: 35625867 PMCID: PMC9138553 DOI: 10.3390/biomedicines10051130] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 02/01/2023] Open
Abstract
Immune checkpoint blockades (ICBs) have revolutionized cancer treatment. Recent studies have revealed a subset of triple-negative breast cancer (TNBC) to be considered as an immunogenic breast cancer subtype. Characteristics of TNBC, such as higher mutation rates and number of tumor-infiltrating immune cells, render the immunogenic phenotypes. Consequently, TNBCs have shown durable responses to ICBs such as atezolizumab and pembrolizumab in clinic. However, a significant number of TNBC patients do not benefit from these therapies, and mechanisms of resistance are poorly understood. Here, we review biomarkers that predict the responsiveness of TNBCs to ICB and recent advances in delineating molecular mechanisms of resistance to ICBs.
Collapse
|
262
|
Zhou Y, Yang J. Narrative review of current status and recommendations in treatment for advanced triple-negative breast cancer. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2022; 3:17. [PMID: 38751541 PMCID: PMC11093051 DOI: 10.21037/tbcr-22-12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/12/2022] [Indexed: 05/18/2024]
Abstract
Background and Objective The encouraging results of clinical trials for triple-negative breast cancer (TNBC) patients in recent years have gradually formed a system for the therapeutic regimen. As immunotherapy and precision therapy based on biomarkers have brought a new era to TNBC, it has become more crucial for clinicians to update the recommended treatment regimens for advanced TNBC patients. Methods We searched literatures related to the treatment for advanced TNBC from 2014 to 2022 on PubMed and sorted out them. In addition, the 2021-2022 guidelines in the part of treatment for advanced TNBC were downloaded on the official website of the guidelines referred to, which were summarized meanwhile. Key Content and Findings We summarized the current status of treatment for advanced TNBC from four aspects: immunotherapy, targeted therapy, antibody-drug conjugates (ADC) drug therapy and chemotherapy. The recommended therapeutic principles for the aspect of advanced TNBC in 2022 Chinese Society of Clinical Oncology (CSCO) guidelines were proposed in combination with international guidelines and conference recommendations. Conclusions It was emphasized that the detection and evaluation of biomarkers or targeting molecules should be the basis for choosing therapeutic regimens in this review and clinicians are supposed to accurately screen the population to achieve better treatment outcomes and prognosis for patients with advanced TNBC.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
263
|
Tan Q, Yin S, Zhou D, Chi Y, Man X, Li H. Potential Predictive and Prognostic Value of Biomarkers Related to Immune Checkpoint Inhibitor Therapy of Triple-Negative Breast Cancer. Front Oncol 2022; 12:779786. [PMID: 35646659 PMCID: PMC9134495 DOI: 10.3389/fonc.2022.779786] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
As an aggressive subtype of breast cancer, triple-negative breast cancer (TNBC) is associated with poor prognosis and lack of effective therapy, except chemotherapy. In recent years, immunotherapy based on immune checkpoint (IC) inhibition has emerged as a promising therapeutic strategy in TNBC. TNBC has more tumor-infiltrating lymphocytes (TILs) and higher rate of mutation and programmed cell death ligand-1 (PD-L1) expression than other subtypes of breast cancer have. However, previous studies have shown that monotherapy has little efficacy and only some TNBC patients can benefit from immunotherapy. Therefore, it is important to identify biomarkers that can predict the efficacy of IC inhibitors (ICIs) in TNBC. Recently, various biomarkers have been extensively explored, such as PD-L1, TILs and tumor mutational burden (TMB). Clinical trials have shown that PD-L1-positive patients with advanced TNBC benefit from ICIs plus chemotherapy. However, in patients with early TNBC receiving neoadjuvant therapy, PD-L1 cannot predict the efficacy of ICIs. These inconsistent conclusions suggest that PD-L1 is the best to date but an imperfect predictive biomarker for efficacy of ICIs. Other studies have shown that advanced TNBC patients with TMB ≥10 mutations/Mb can achieve clinical benefits from pembrolizumab. TILs also have potential predictive value in TNBC. Here, we select some biomarkers related to ICIs and discuss their potential predictive and prognostic value in TNBC. We hope these biomarkers could help to identify suitable patients and realize precision immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Huihui Li
- Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
264
|
Rizzo A, Cusmai A, Gadaleta-Caldarola G, Palmiotti G. Current and Future Role of Neoadjuvant Chemoimmunotherapy for Early Triple-Negative Breast Cancer: Which Way to Go Forward. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:600. [PMID: 35630016 PMCID: PMC9146481 DOI: 10.3390/medicina58050600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022]
Abstract
Immunotherapy has revolutionized previous triple-negative breast cancer (TNBC) treatment algorithms, prompting researchers and clinicians to consider the expansion of the role of immunotherapy in other settings, including the earlier stage of the disease (e.g., as neoadjuvant and adjuvant therapy). The role of chemoimmunotherapy have been assessed in some recently presented and published clinical trials, including the KEYNOTE-522, the IMpassion031, and the GeparNUEVO. In the current Editorial, we will provide a critical snapshot of these studies, exploring strengths and limitations of neoadjuvant chemotherapy in early TNBC.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (G.P.)
| | - Antonio Cusmai
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (G.P.)
| | - Gennaro Gadaleta-Caldarola
- Medical Oncology Unit, ‘Mons. R. Dimiccoli’ Hospital, Azienda Sanitaria Locale Barletta, 76121 Barletta, Italy;
| | - Gennaro Palmiotti
- Struttura Semplice Dipartimentale di Oncologia Medica per la Presa in Carico Globale del Paziente Oncologico “Don Tonino Bello”, I.R.C.C.S. Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy; (A.C.); (G.P.)
| |
Collapse
|
265
|
Muhammad A, Forcados GE, Katsayal BS, Bako RS, Aminu S, Sadiq IZ, Abubakar MB, Yusuf AP, Malami I, Faruk M, Ibrahim S, Pase PA, Ahmed S, Abubakar IB, Abubakar M, Yates C. Potential epigenetic modifications implicated in triple- to quadruple-negative breast cancer transition: a review. Epigenomics 2022; 14:711-726. [PMID: 35473304 DOI: 10.2217/epi-2022-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Current research on triple-negative breast cancer (TNBC) has resulted in delineation into the quadruple-negative breast cancer (QNBC) subgroup. Epigenetic modifications such as DNA methylation, histone posttranslational modifications and associated changes in chromatin architecture have been implicated in breast cancer pathogenesis. Herein, the authors highlight genes with observed epigenetic modifications that are associated with more aggressive TNBC/QNBC pathogenesis and possible interventions. Advanced literature searches were done on PubMed/MEDLINE, Scopus and Google Scholar. The results suggest that nine epigenetically altered genes/differentially expressed proteins in addition to the downregulated androgen receptor are associated with TNBC aggressiveness and could be implicated in the TNBC to QNBC transition. Thus, restoring the normal expression of these genes via epigenetic reprogramming could be therapeutically beneficial to TNBC and QNBC patients.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria.,Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| | | | - Babangida Sanusi Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Rabiatu Suleiman Bako
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Suleiman Aminu
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Idris Zubairu Sadiq
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Murtala Bello Abubakar
- Department of Physiology, Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Sokoto State, Nigeria.,Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Sokoto State, Nigeria
| | | | - Ibrahim Malami
- Department of Pharmacognosy & Ethnopharmacy, Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Nigeria.,Centre for Advanced Medical Research & Training (CAMRET), Usmanu Danfodiyo University, P.M.B 2254, Sokoto, Sokoto State, Nigeria
| | - Mohammed Faruk
- Department of Pathology, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Sani Ibrahim
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Peter Abur Pase
- Department of Surgery, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Saad Ahmed
- Department of Pathology, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Ibrahim Babangida Abubakar
- Deparment of Biochemistry, Kebbi State University of Science & Technology, PMB 1144, Aliero, Kebbi State, Nigeria
| | - Murtala Abubakar
- Department of Pathology, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Clayton Yates
- Center for Cancer Research, Department of Biology, Tuskegee University, Tuskegee, AL 36088, USA
| |
Collapse
|
266
|
Agostinetto E, Montemurro F, Puglisi F, Criscitiello C, Bianchini G, Del Mastro L, Introna M, Tondini C, Santoro A, Zambelli A. Immunotherapy for HER2-Positive Breast Cancer: Clinical Evidence and Future Perspectives. Cancers (Basel) 2022; 14:2136. [PMID: 35565264 PMCID: PMC9105460 DOI: 10.3390/cancers14092136] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common malignancy among women worldwide, and HER2-positive breast cancer accounts for approximately 15% of all breast cancer diagnoses. The advent of HER2-targeting therapies has dramatically improved the survival of these patients, significantly reducing their risk of recurrence and death. However, as a significant proportion of patients ultimately develop resistance to these therapies, it is extremely important to identify new treatments to further improve their clinical outcomes. Immunotherapy has revolutionized the treatment and history of several cancer types, and it has already been approved as a standard of care for patients with triple-negative breast cancer. Based on a strong preclinical rationale, immunotherapy in HER2-positive breast cancer represents an intriguing field that is currently under clinical investigation. There is a close interplay between HER2-targeting therapies (both approved and under investigation) and the immune system, and several new immunotherapeutic strategies, including immune checkpoint inhibitors, CAR-T cells and therapeutic vaccines, are being studied in this disease. In this narrative review, we discuss the clinical evidence and the future perspectives of immunotherapy for patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B), 1070 Brussels, Belgium;
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Filippo Montemurro
- Direzione Breast Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Fabio Puglisi
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy;
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development, European Institute of Oncology IRCCS, 20141 Milan, Italy;
- Department of Oncology and Hematology, University of Milan, 20122 Milan, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Lucia Del Mastro
- IRCCS Ospedale Policlinico San Martino, Clinica di Oncologia Medica, 16132 Genova, Italy;
- Dipartimento di Medicina Interna e Specialità Medica, Università di Genova, 16124 Genova, Italy
| | - Martino Introna
- UOS Centro di Terapia Cellulare “G. Lanzani”, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy;
| | - Carlo Tondini
- Medical Oncology Unit, ASST Papa Giovanni XXIII, Piazza OMS 1, 27100 Bergamo, Italy;
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
267
|
Wang M, Feng R, Chen Z, Shi W, Li C, Liu H, Wu K, Li D, Li X. Identification of Cancer-Associated Fibroblast Subtype of Triple-Negative Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6452636. [PMID: 35505821 PMCID: PMC9057104 DOI: 10.1155/2022/6452636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND There is limited knowledge about the role of cancer-associated fibroblasts (CAF) in the tumor microenvironment of triple-negative breast cancer (TNBC). METHODS Three hundred and thirty-five TNBC samples from four datasets were retrieved and analyzed. In order to determine the CAF subtype by combining gene expression profiles, an unsupervised clustering analysis was adopted. The prognosis, enriched pathways, immune cells, immune scores, and tumor purity were compared between CAF subtypes. The genes with the highest importance were selected by bioinformatics analysis. The machine learning model was built to predict the TNBC CAF subtype by these selected genes. RESULTS TNBC samples were classified into two CAF subtypes (CAF+ and CAF-). The CAF- subtype of TNBC was linked to the longer overall survival and more immune cells than the CAF+ subtype. CAF- and CAF+ were enriched in immune-related pathways and extracellular matrix pathways, respectively. Bioinformatics analysis identified 9 CAF subtype-related markers (ADAMTS12, AEBP1, COL10A1, COL11A1, CXCL11, CXCR6, EDNRA, EPPK1, and WNT7B). We constructed a robust random forest model using these 9 genes, and the area under the curve (AUC) value of the model was 0.921. CONCLUSION The current study identified CAF subtypes based on gene expression profiles and found that CAF subtypes have significantly different overall survival, immune cells, and immunotherapy response rates.
Collapse
Affiliation(s)
- Maoli Wang
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Ruifa Feng
- Breast Center of the Second Affiliated Hospital of Guilin Medical University, 541100 Guilin, Guangxi, China
| | - Zihao Chen
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, 26121 Oldenburg, Germany
| | - Wenjie Shi
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, 26121 Oldenburg, Germany
| | - Cheng Li
- Department of Orthopaedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, 100035 Beijing, China
| | - Huiquan Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, 230032 Hefei, China
| | - Kejin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Dajin Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, China
| | - Xiqing Li
- Oncology Department, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, 450003 Zhengzhou, China
| |
Collapse
|
268
|
Carlino F, Diana A, Piccolo A, Ventriglia A, Bruno V, De Santo I, Letizia O, De Vita F, Daniele B, Ciardiello F, Orditura M. Immune-Based Therapy in Triple-Negative Breast Cancer: From Molecular Biology to Clinical Practice. Cancers (Basel) 2022; 14:cancers14092102. [PMID: 35565233 PMCID: PMC9103968 DOI: 10.3390/cancers14092102] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has been considered for many years an orphan disease in terms of therapeutic options, with conventional chemotherapy (CT) still representing the mainstay of treatment in the majority of patients. Although breast cancer (BC) has been historically considered a "cold tumor", exciting progress in the genomic field leading to the characterization of the molecular portrait and the immune profile of TNBC has opened the door to novel therapeutic strategies, including Immune Checkpoint Inhibitors (ICIs), Poly ADP-Ribose Polymerase (PARP) inhibitors and Antibody Drug Conjugates (ADCs). In particular, compared to standard CT, the immune-based approach has been demonstrated to improve progression-free survival (PFS) and overall survival (OS) in metastatic PD-L1-positive TNBC and the pathological complete response rate in the early setting, regardless of PD-L1 expression. To date, PD-L1 has been widely used as a predictor of the response to ICIs; however, many patients do not benefit from the addition of immunotherapy. Therefore, PD-L1 is not a reliable predictive biomarker of the response, and its accuracy remains controversial due to the lack of a consensus about the assay, the antibody, and the scoring system to adopt, as well as the spatial and temporal heterogeneity of the PD-L1 status. In the precision medicine era, there is an urgent need to identify more sensitive biomarkers in the BC immune oncology field other than just PD-L1 expression. Through the characterization of the tumor microenvironment (TME), the analysis of peripheral blood and the evaluation of immune gene signatures, novel potential biomarkers have been explored, such as the Tumor Mutational Burden (TMB), Microsatellite Instability/Mismatch Repair Deficiency (MSI/dMMR) status, genomic and epigenomic alterations and tumor-infiltrating lymphocytes (TILs). This review aims to summarize the recent knowledge on BC immunograms and on the biomarkers proposed to support ICI-based therapy in TNBC, as well as to provide an overview of the potential strategies to enhance the immune response in order to overcome the mechanisms of resistance.
Collapse
Affiliation(s)
- Francesca Carlino
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
- Medical Oncology Unit, Ospedale Ave Gratia Plena, San Felice a Cancello, 81027 Caserta, Italy; (I.D.S.); (O.L.)
- Correspondence: ; Tel.: +39-349-5152216
| | - Anna Diana
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (A.D.); (B.D.)
| | - Antonio Piccolo
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Anna Ventriglia
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Vincenzo Bruno
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Irene De Santo
- Medical Oncology Unit, Ospedale Ave Gratia Plena, San Felice a Cancello, 81027 Caserta, Italy; (I.D.S.); (O.L.)
| | - Ortensio Letizia
- Medical Oncology Unit, Ospedale Ave Gratia Plena, San Felice a Cancello, 81027 Caserta, Italy; (I.D.S.); (O.L.)
| | - Ferdinando De Vita
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Bruno Daniele
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (A.D.); (B.D.)
| | - Fortunato Ciardiello
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| | - Michele Orditura
- Department of Precision Medicine, Division of Medical Oncology, University of Campania Luigi Vanvitelli, 80131 Naples, Italy; (A.P.); (A.V.); (V.B.); (F.D.V.); (F.C.); (M.O.)
| |
Collapse
|
269
|
Almario EN, Kettermann A, Popat V. Informatics in Medical Product Regulation: The Right Drug at the Right Dose for the Right Patient. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2486:277-314. [PMID: 35437728 DOI: 10.1007/978-1-0716-2265-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This chapter describes the role of regulatory medical product review in furthering precision medicine. Efficient data processing and appropriate analyses are needed to synthesize information and provide directions for use in a medical product label. We describe opportunities and challenges in outcome assessment through informatics, as bioengineered therapeutics are increasingly developed for the unmet needs of molecularly defined diseases. Data submission requirements and analytic principles are outlined, and regulatory resources and foundational law and statute are cited for the reader.
Collapse
Affiliation(s)
| | - Anna Kettermann
- Office of Biostatistics, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Vaishali Popat
- Biomedical Informatics and Regulatory Review Science, Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
270
|
Szeitz B, Pipek O, Kulka J, Szundi C, Rusz O, Tőkés T, Szász AM, Kovács KA, Pesti A, Ben Arie TB, Gángó A, Fülöp Z, Drágus E, Vári-Kakas SA, Tőkés AM. Investigating the Prognostic Relevance of Tumor Immune Microenvironment and Immune Gene Assembly in Breast Carcinoma Subtypes. Cancers (Basel) 2022; 14:1942. [PMID: 35454849 PMCID: PMC9031175 DOI: 10.3390/cancers14081942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/29/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
We hypothesized that different BC subtypes are characterized by spatially distinct tumor immune microenvironment (TIME) and that immune gene assembly of metastatic (Met) and non-metastatic (Ctrl) BCs vary across subtypes. Peritumoral, stromal and intratumoral TIL was assessed on 309 BC cases. Hot, cold and immune-excluded groups were defined, and the prognostic role of this classification was assessed. CD4+/CD8+ positivity was analyzed in 75 cases in four systematically predefined tumor regions. Immune gene expression of Met and Ctrl HER2-negative BCs was compared by using NanoString nCounter technology. The amount of TIL infiltration varied greatly within all BC subtypes. Two-third of the cases were cold tumors with no significant survival difference compared to hot tumors. A lower CD4+/CD8+ ratio at the stromal internal tumor region was significantly associated with longer distant metastasis-free survival. The differentially expressed immune genes between Met and Ctrl varied across the studied BC subtypes with TNBC showing distinct features from the luminal subtypes. The TIME is characterized by a considerable heterogeneity; however, low level of TILs does not equate to disease progression. The differences in immune gene expression observed between Met and Ctrl breast carcinomas call attention to the important role of altered immune function in BC progression.
Collapse
Affiliation(s)
- Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.); (T.T.); (A.M.S.)
| | - Orsolya Pipek
- Department of Physics of Complex Systems, Institute of Physics, Eötvös Loránd University, 1117 Budapest, Hungary;
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary; (J.K.); (C.S.); (K.A.K.); (A.P.); (T.B.B.A.)
| | - Csilla Szundi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary; (J.K.); (C.S.); (K.A.K.); (A.P.); (T.B.B.A.)
| | - Orsolya Rusz
- Department of Pathology, Forensic and Insurance Medicine, SE NAP, Brain Metastasis Research Group, Semmelweis University, 1091 Budapest, Hungary;
| | - Tímea Tőkés
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.); (T.T.); (A.M.S.)
| | - Attila Marcell Szász
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1083 Budapest, Hungary; (B.S.); (T.T.); (A.M.S.)
- Department of Bioinformatics, Semmelweis University, 1052 Budapest, Hungary
| | - Kristóf Attila Kovács
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary; (J.K.); (C.S.); (K.A.K.); (A.P.); (T.B.B.A.)
| | - Adrián Pesti
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary; (J.K.); (C.S.); (K.A.K.); (A.P.); (T.B.B.A.)
| | - Taya Beri Ben Arie
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary; (J.K.); (C.S.); (K.A.K.); (A.P.); (T.B.B.A.)
| | - Ambrus Gángó
- MTA-SE Momentum Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary;
| | - Zsolt Fülöp
- Department of Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology, 540139 Targu Mures, Romania;
| | - Emőke Drágus
- Department of Urology, Clinical County Hospital, 540167 Targu Mures, Romania;
| | - Stefan A. Vári-Kakas
- Faculty of Electrical Engineering and Information Technology, University of Oradea, Str. Universitatii nr. 1, 410087 Oradea, Romania;
| | - Anna Mária Tőkés
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, 1091 Budapest, Hungary; (J.K.); (C.S.); (K.A.K.); (A.P.); (T.B.B.A.)
| |
Collapse
|
271
|
Huppert LA, Gumusay O, Rugo HS. Emerging treatment strategies for metastatic triple-negative breast cancer. Ther Adv Med Oncol 2022; 14:17588359221086916. [PMID: 35422881 PMCID: PMC9003656 DOI: 10.1177/17588359221086916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/23/2022] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous subtype of breast cancer that is often associated with an aggressive phenotype and a poor prognosis. Cytotoxic chemotherapy remains the mainstay of treatment for most patients with metastatic TNBC (mTNBC), but duration of response is often short and median overall survival is only 12-18 months. Therefore, it is critical to identify novel treatment strategies to improve outcomes for these patients. In this review article, we discuss recent advances in treatment strategies for patients with mTNBC including the use of immune checkpoint inhibitors, targeted therapies, and antibody-drug conjugates. For each topic, we summarize important preclinical and clinical data, discuss implications for clinical practice, and highlight future research directions.
Collapse
Affiliation(s)
- Laura A. Huppert
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Ozge Gumusay
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Hope S. Rugo
- Director, Breast Oncology and Clinical Trials Education, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1600 Divisadero St., Box 1710, San Francisco, CA 94115, USA
| |
Collapse
|
272
|
Li Q, Zhong X, Yao W, Yu J, Wang C, Li Z, Lai S, Qu F, Fu X, Huang X, Zhang D, Liu Y, Li H. Inhibitor of glutamine metabolism V9302 promotes ROS-induced autophagic degradation of B7H3 to enhance antitumor immunity. J Biol Chem 2022; 298:101753. [PMID: 35189139 PMCID: PMC8968643 DOI: 10.1016/j.jbc.2022.101753] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
Despite the enormous successes of anti-PD-1/PD-L1 immunotherapy in multiple other cancer types, the overall response rates of breast cancer remain suboptimal. Therefore, exploring additional immune checkpoint molecules for potential cancer treatment is crucial. B7H3, a T-cell coinhibitory molecule, is specifically overexpressed in breast cancer compared with normal breast tissue and benign lesions, making it an attractive therapeutic target. However, the mechanism by which B7H3 contributes to the cancer phenotype is unclear. Here we show that the expression of B7H3 is negatively related to the number of CD8+ T cells in breast tumor sites. In addition, analysis of the differentially expressed B7H3 reveals that it is inversely correlated to autophagic flux both in breast cancer cell lines and clinical tumor tissues. Furthermore, block of autophagy by bafilomycin A1 (Baf A1) increases B7H3 levels and attenuates CD8+ T cell activation, while promotion of autophagy by V9302, a small-molecule inhibitor of glutamine metabolism, decreases B7H3 expression and enhances granzyme B (GzB) production of CD8+ T cells via regulation of reactive oxygen species (ROS) accumulation. We demonstrate that combined treatment with V9302 and anti-PD-1 monoclonal antibody (mAb) enhances antitumor immunity in syngeneic mouse models. Collectively, our findings unveil the beneficial effect of V9302 in boosting antitumor immune response in breast cancer and illustrate that anti-PD-1 together with V9302 treatment may provide synergistic effects in the treatment of patients insensitive to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Qian Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaofang Zhong
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weicheng Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junli Yu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chao Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zongyan Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Hepatobiliary Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shengqing Lai
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fanli Qu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Fu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojia Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dawei Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Hepatobiliary Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Yujie Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, 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.
| | - Haiyan Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Breast Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
273
|
Corti C, Venetis K, Sajjadi E, Zattoni L, Curigliano G, Fusco N. CAR-T cell therapy for triple-negative breast cancer and other solid tumors: preclinical and clinical progress. Expert Opin Investig Drugs 2022; 31:593-605. [PMID: 35311430 DOI: 10.1080/13543784.2022.2054326] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Most breast cancer-related deaths arise from triple-negative breast cancer (TNBC). Molecular heterogeneity, aggressiveness and the lack of effective therapies are major hurdles to therapeutic progress. Chimeric antigen receptor (CAR)-T cells have emerged as a promising immunotherapeutic strategy in TNBC. This approach combines the antigen specificity of an antibody with the effector function of T cells. AREAS COVERED This review examines the opportunities provided by CAR-T cell therapies in solid tumors. Emerging targets, ongoing clinical trials, and prospective clinical implications in TNBC are considered later. An emphasis is placed on the key challenges and possible solutions for this therapeutic approach. EXPERT OPINION A challenge for CAR-T cell therapy is the selection of the optimal targets to minimize on-target/off-tumor toxicity. Tumor escape via antigen loss and intrinsic heterogeneity is a further hurdle. TROP2, GD2, ROR1, MUC1 and EpCAM are promising targets. Persistence and trafficking to tumor cells may be enhanced by the implementation of CARs with a chemokine receptor and/or constitutively activated interleukin receptors. Fourth-generation CARs (TRUCKs) may redirect T-cells for universal cytokine-mediated killing. Combinatorial approaches and the application of CARs to other immune cells could revert the suppressive immune environment that characterizes solid neoplasms.
Collapse
Affiliation(s)
- Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Lorenzo Zattoni
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
274
|
Elliott MJ, Wilson B, Cescon DW. Current Treatment and Future Trends of Immunotherapy in Breast Cancer. Curr Cancer Drug Targets 2022; 22:667-677. [PMID: 35301950 DOI: 10.2174/1568009622666220317091723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022]
Abstract
Immunotherapy continues to redefine the solid tumor treatment landscape with inhibitors of the PD-L1/PD-1 immune checkpoint having the most widespread impact. As the most common cancer diagnosed worldwide, there is significant interest in the development of immunotherapy for the treatment of breast cancer in both the early and metastatic settings. Recently reported results of several clinical trials have identified potential roles for immunotherapy agents alone or in combination with standard treatment for early and metastatic disease. While trials to date have been promising, immunotherapy thus far has been shown to benefit only a select group of patients with breast cancer, defined by tumor subtype, PD-L1 expression, and line of therapy. With over 250 trials ongoing, emerging data will enable the further refinement of breast cancer immunotherapy strategies. The integration of multiple putative biomarkers and consideration of dynamic markers of early response or resistance may inform optimal patient selection for immunotherapy investigation and integration into clinical practice. This review will summarize the current evidence for immune-checkpoint blockade (ICB) in the treatment of early and metastatic breast cancer and highlight current and potential future biomarkers of therapeutic response.
Collapse
Affiliation(s)
- Mitchell J Elliott
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Brooke Wilson
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
- University of New South Wales, Kensington, New South Wales, Australia
| | - David W Cescon
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
275
|
Tabernero J, Andre F, Blay JY, Bustillos A, Fear S, Ganta S, Jaeger D, Maio M, Mileshkin L, Melero I. Phase II multicohort study of atezolizumab monotherapy in multiple advanced solid cancers. ESMO Open 2022; 7:100419. [PMID: 35305400 PMCID: PMC9058880 DOI: 10.1016/j.esmoop.2022.100419] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The programmed death-ligand 1 inhibitor atezolizumab had shown clinical activity against several advanced malignancies. PATIENTS AND METHODS This phase II, open-label basket study (NCT02458638) was conducted in 16 main cohorts of patients aged ≥18 years with stage III or IV solid tumors. In stage I, 12 patients were enrolled into each cohort. Treatment was atezolizumab 1200 mg intravenously every 3 weeks until loss of clinical benefit or unacceptable toxicity. The primary efficacy endpoint was the non-progression rate (NPR) at 18 weeks in treated, assessable patients. NPR ≤20% was not of interest for development as monotherapy, and NPR ≥40% was defined as the threshold of benefit/success. If ≥3 patients had non-progressive disease in stage I (interim analysis), 13 additional patients could be enrolled into stage II (final analysis). Secondary efficacy and safety endpoints were also evaluated. RESULTS Overall, 474 patients were enrolled and treated; 433 were included in the efficacy set. Due partly to slow recruitment because of competing trials and limited efficacy at interim analyses, enrollment was stopped early, including in cohorts that passed stage I boundaries of success. NPR was >20% in five cohorts: cervical cancer {n = 27; NPR 44.4% [95% confidence interval (CI) 25.5% to 64.7%]}; follicular/papillary thyroid cancer [n = 11; 54.5% (95% CI 23.4% to 83.3%)]; thymoma [n = 13; 76.9% (95% CI: 46.2% to 95.0%)]; gastroenteropancreatic (GEP) and lung neuroendocrine tumors [NETs; n = 24; 41.7% (95% CI 22.1% to 63.4%)], and low/intermediate grade carcinoid GEP and lung NETs [n = 12; 58.3% (95% CI 27.7% to 84.8%)]. Treatment-related adverse events occurred in 55.3% of patients overall, and at grade 3, 4, and 5 in 10.3%, 1.7%, and 0.4%, respectively. CONCLUSIONS Atezolizumab monotherapy was effective in the cervical cancer cohort. The interim benefit threshold was crossed in patients with follicular/papillary thyroid cancer, thymoma, and GEP and lung NETs, but recruitment was stopped before these signals could be confirmed in stage II. Safety was consistent with previous findings.
Collapse
Affiliation(s)
- J Tabernero
- Medical Oncology Department - Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), UVic-UCC, IOB-Quiron, Barcelona, Spain.
| | - F Andre
- Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - J-Y Blay
- Centre Léon Bérard, Lyon, France
| | - A Bustillos
- Global Product Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - S Fear
- Global Product Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - S Ganta
- Product Development Safety, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - D Jaeger
- Medical Oncology Department, Heidelberg University Hospital (UKHD), Heidelberg, Germany
| | - M Maio
- Center for Immuno-Oncology, University Hospital of Siena, Siena, Italy
| | - L Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - I Melero
- Centre of Applied Medical Research, University Clinic of Navarra, Navarra, Spain
| |
Collapse
|
276
|
Magbanua MJM, Gumusay O, Kurzrock R, van ‘t Veer LJ, Rugo HS. Immunotherapy in Breast Cancer and the Potential Role of Liquid Biopsy. Front Oncol 2022; 12:802579. [PMID: 35372077 PMCID: PMC8964955 DOI: 10.3389/fonc.2022.802579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Liquid biopsy biomarkers, such as circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), are noninvasive diagnostics that could complement predictive and prognostic tools currently used in the clinic. Recent trials of immunotherapy have shown promise in improving outcomes in a subset of breast cancer patients. Biomarkers could improve the efficacy of immune checkpoint inhibitors by identifying patients whose cancers are more likely to respond to immunotherapy. In this review, we discuss the current applications of liquid biopsy and emerging technologies for evaluation of immunotherapy response and outcomes in breast cancer. We also provide an overview of the status of immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Mark Jesus M. Magbanua
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Ozge Gumusay
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, United States
| | - Razelle Kurzrock
- Worldwide Innovative Network (WIN) for Personalized Cancer Therapy Consortium, Villejuif, France
| | - Laura J. van ‘t Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Hope S. Rugo
- Division of Hematology Oncology, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
277
|
Yang R, Li Y, Wang H, Qin T, Yin X, Ma X. Therapeutic progress and challenges for triple negative breast cancer: targeted therapy and immunotherapy. MOLECULAR BIOMEDICINE 2022; 3:8. [PMID: 35243562 PMCID: PMC8894518 DOI: 10.1186/s43556-022-00071-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer, with estrogen receptor, human epidermal growth factor receptor 2 and progesterone receptor negative. TNBC is characterized by high heterogeneity, high rates of metastasis, poor prognosis, and lack of therapeutic targets. Now the treatment of TNBC is still based on surgery and chemotherapy, which is effective only in initial stage but almost useless in advanced stage. And due to the lack of hormone target, hormonal therapies have little beneficial effects. In recent years, signaling pathways and receptor-specific targets have been reported to be effective in TNBC patients under specific clinical conditions. Now targeted therapies have been approved for many other cancers and even other subtypes of breast cancer, but treatment options for TNBC are still limited. Most of TNBC patients showed no response, which may be related to the heterogeneity of TNBC, therefore more effective treatments and predictive biomarkers are needed. In the present review, we summarize potential treatment opinions for TNBC based on the dysregulated receptors and signaling pathways, which play a significant role in multiple stages of TNBC development. We also focus on the application of immunotherapy in TNBC, and summarize the preclinical and clinical trials of therapy for patients with TNBC. We hope to accelerate the research and development of new drugs for TNBC by understanding the relevant mechanisms, and to improve survival.
Collapse
Affiliation(s)
- Ruoning Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.,Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yueyi Li
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Hang Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Taolin Qin
- West China Hospital, West China Medical School Sichuan University, Chengdu, PR, China
| | - Xiaomeng Yin
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy,Cancer Center, West China Hospital, 37 Guoxue Alley, Chengdu, 610041, PR, China.
| |
Collapse
|
278
|
Agostinetto E, Losurdo A, Nader-Marta G, Santoro A, Punie K, Barroso R, Popovic L, Solinas C, Kok M, de Azambuja V, Lambertini M. Progress and pitfalls in the use of immunotherapy for patients with triple negative breast cancer. Expert Opin Investig Drugs 2022; 31:567-591. [PMID: 35240902 DOI: 10.1080/13543784.2022.2049232] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Triple negative breast cancer (TNBC) is an area of high unmet medical need in terms of new effective treatment strategies. Although breast cancer is traditionally considered a 'cold' tumor type, TNBC is the most appropriate subtype for immunotherapeutic strategies; this is due to the high level of tumor infiltrating lymphocytes, PD-L1 expression and tumor mutational burden relative to other breast cancer subtypes. AREAS COVERED This review examines the use of immunotherapeutic strategies in early and advanced TNBC. The paper summarizes data on novel promising immunomodulatory approaches that have been explored in early phase trials and discusses the pitfalls and limitations often encountered in clinical research. EXPERT OPINION PD-1-blockade is approved for stage II/III TNBC and for first-line treatment of PD-L1-positive TNBC patients with metastatic disease and should be considered standard of care. However, question marks and difficulties remain; these include the identification of predictive biomarkers to select patients who benefit from the addition of PD1-blockade and the balance between efficacy and long-term toxicity for an individual patient. Numerous treatment combinations and new immunotherapeutic strategies beyond PD1 blockade are being evaluated, thus reflecting a promising evolution of a more personalized approach, and extended clinical benefit in TNBC.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium.,Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Agnese Losurdo
- Humanitas Research Hospital - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.,Humanitas Research Hospital - IRCCS, Humanitas Cancer Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | | | - Lazar Popovic
- Oncology Institute of Vojvodina, Faculty of Medicine, University Novi Sad, Novi Sad, Serbia
| | - Cinzia Solinas
- Medical Oncology, ATS Sardegna, Ospedale San Francesco, Nuoro, Italy
| | - Marleen Kok
- Departments of Medical Oncology, Tumor Biology & Immunology. Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Vandro de Azambuja
- Academic Trials Promoting Team, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
279
|
Hall PE, Schmid P. Emerging strategies for TNBC with early clinical data: new chemoimmunotherapy strategies. Breast Cancer Res Treat 2022; 193:21-35. [PMID: 35235095 DOI: 10.1007/s10549-022-06547-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 02/20/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE The use of immune checkpoint inhibitors in combination with chemotherapy for the treatment of triple-negative breast cancer is becoming more widespread as efficacy data accumulates. However, outcomes remain less than optimal and not all patients benefit from treatment. The aim of this article is to review the emerging chemoimmunotherapy strategies for triple-negative breast cancer. METHODS Searches were undertaken on Pubmed and Clinicaltrials.gov for relevant publications and trials. RESULTS Preclinical and clinical data have provided insights into the differing immunomodulatory effects of chemotherapy agents, highlighting the immunostimulatory properties of anthracyclines. Mechanisms of resistance to immune checkpoint inhibition are discussed and the potential role of phosphatidylinositol 3-kinase (PI3K)/AKT and MAPK/ERK kinase (MEK) inhibitors in overcoming resistance. Finally, the emerging therapeutic class of antibody-drug conjugates for triple-negative breast cancer in combination with immune checkpoint inhibitors is reviewed. CONCLUSIONS The type and sequence of chemotherapy agents play an important role in optimising the response to immune checkpoint inhibitors. Antibody-drug conjugates in combination with immune checkpoint inhibitors are a promising area of development.
Collapse
Affiliation(s)
- Peter E Hall
- Department of Medical Oncology, St Bartholomew's Hospital, King George V Building, West Smithfield, London, EC1A 7BE, UK.
| | - Peter Schmid
- Department of Medical Oncology, St Bartholomew's Hospital, King George V Building, West Smithfield, London, EC1A 7BE, UK.,Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
280
|
Zheng H, Guo B, Qiu X, Xia Y, Qu Y, Cheng L, Meng F, Zhong Z. Polymersome-mediated cytosolic delivery of cyclic dinucleotide STING agonist enhances tumor immunotherapy. Bioact Mater 2022; 16:1-11. [PMID: 35386324 PMCID: PMC8958419 DOI: 10.1016/j.bioactmat.2022.02.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 12/16/2022] Open
Abstract
Cyclic dinucleotides (CDNs) as stimulator of interferon genes (STING) agonists capable of inducing strong antitumor innate immune response are highly promising for tumor immunotherapy. The efficacy of these CDNs is, however, reduced greatly by their fast clearance, poor cell uptake and inefficient cytosolic transportation. Here, we report that reduction-responsive biodegradable chimaeric polymersomes (CPs) markedly enhance tumor retention and cytosolic delivery of a synthetic CDN, ADU-S100, and bolster STING pathway activation in the tumor microenvironment and tumor draining lymph nodes, giving significantly better tumor repression and survival of B16F10 melanoma-bearing mice compared with free CDN control. The superiority of CPs-mediated CDN delivery is further verified in combination therapy with low-dose fractionated radiation, which brings about clearly stronger and longer-term immunotherapeutic effects and protection against tumor re-challenge. The development of nano-STING agonists that are able to overcome the delivery barriers of CDNs represents an effective strategy to potentiate cancer immunotherapy. Chimaeric polymersomes (CPs) show efficient loading of STING agonist cyclic dinucleotide. Intratumoral injection of CPs-CDN gives notable tumor accumulation and cytosolic delivery of STING agonist. CPs-CDN activate STING pathway in tumor microenvironment and tumor draining lymph node, giving significant tumor repression and survival of melanoma mice. CPs-CDN combined with low-dose fractionated X ray irradiation led to further boosted immunotherapy with 3/7 cured mice. Nano-STING agonists can overcome the delivery barriers of CDNs and potentiate durable cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Zheng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Xinyun Qiu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Yifeng Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Yan Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Liang Cheng
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
- Corresponding author.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- Corresponding author.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
- Corresponding author. Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China.
| |
Collapse
|
281
|
Tumor Cell-Autonomous Pro-Metastatic Activities of PD-L1 in Human Breast Cancer Are Mediated by PD-L1-S283 and Chemokine Axes. Cancers (Basel) 2022; 14:cancers14041042. [PMID: 35205789 PMCID: PMC8870053 DOI: 10.3390/cancers14041042] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Triple-negative breast cancer (TNBC) is an aggressive disease that responds in a limited manner to immune checkpoint blockades targeting the PD-L1/PD-1 axis, suggesting that PD-L1 potentiates TNBC progression via pathways not related to immune suppression. We demonstrated that, in human breast cancer cells, PD-L1 expression increased in a cell-autonomous manner tumor cell growth, invasion and release of pro-metastatic factors; these activities were elevated by exposure to PD-1 and were markedly impaired in S283-mutated PD-L1-expressing cells. Invasion of WT-PD-L1-expressing TNBC cells depended on autocrine chemokine circuits, involving CXCR1/2, CCR2, CCR5 and their ligands. In T cell-deficient mice, WT-PD-L1 exhibited increased tumor growth and metastasis by TNBC cells, whereas S283A-PD-L1-expressing cells showed a very poor tumorigenic and metastatic profile. These findings on cell-autonomous and PD-1-induced pro-metastatic activities of PD-L1 in cancer cells suggest that treatments targeting PD-L1 could improve the efficacy of immune-targeting checkpoint inhibitors, e.g., anti-PD-1 or anti-CTLA-4 in TNBC. Abstract Therapies targeting the PD-L1/PD-1 axis have recently been introduced to triple-negative breast cancer (TNBC) with limited efficacy, suggesting that this axis promotes tumor progression through mechanisms other than immune suppression. Here, we over-expressed WT-PD-L1 in human TNBC cells (express endogenous PD-L1) and in luminal-A breast cancer cells (no endogenous PD-L1 expression) and demonstrated that cell-autonomous PD-L1 activities lead to increased tumor cell growth, invasion and release of pro-metastatic factors (CXCL8, sICAM-1, GM-CSF). These activities were promoted by PD-1 and were inhibited by mutating S283 in PD-L1. Invasion of WT-PD-L1-cells required signaling by chemokine receptors CXCR1/2, CCR2 and CCR5 through autocrine circuits involving CXCL8, CCL2 and CCL5. Studies with T cell-deficient mice demonstrated that cell-autonomous WT-PD-L1 activities in TNBC cells increased tumor growth and metastasis compared to knock-out (KO)-PD-L1-cells, whereas S283A-PD-L1-expressing cells had minimal ability to form tumors and did not metastasize. Overall, our findings reveal autonomous and PD-1-induced tumor-promoting activities of PD-L1 that depend on S283 and on chemokine circuits. These results suggest that TNBC patients whose tumors express PD-L1 could benefit from therapies that prevent immune suppression by targeting PD-1/CTLA-4, alongside with antibodies to PD-L1, which would allow maximal impact by mainly targeting the cancer cells.
Collapse
|
282
|
Wang C, Feng G, Zhu J, Wei K, Huang C, Wu Z, Yu Y, Qin G. Developing an immune signature for triple-negative breast cancer to predict prognosis and immune checkpoint inhibitor response. Future Oncol 2022; 18:1055-1066. [PMID: 35105171 DOI: 10.2217/fon-2021-0600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We aimed to develop a new signature based on immune-related genes to predict prognosis and response to immune checkpoint inhibitors in patients with triple-negative breast cancer (TNBC). Materials & methods: Single-sample gene set enrichment was used to develop an immune-based prognostic signature (IPRS) for TNBC patients. We conducted multivariate Cox analysis to evaluate the prognosis value of the IPRS. Result: An IPRS based on 66 prognostic genes was developed. Multivariate Cox analysis indicated that the IPRS was an independent factor for prognosis. PD-1, PD-L1, PD-L2 and CTLA4 gene expression was higher in the low-risk group, suggesting IPRS could predict the response to immune checkpoint inhibitors. Conclusion: The IPRS might be a reliable signature to predict TNBC patients' prognosis and response to immune checkpoint inhibitors, but needs prospective validation.
Collapse
Affiliation(s)
- Ce Wang
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100083, China
| | - Guoshuang Feng
- Big Data & Engineering Research Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100083, China
| | - Jingjing Zhu
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Kecheng Wei
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Chen Huang
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Zhenyu Wu
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Yongfu Yu
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China
| | - Guoyou Qin
- Department of Biostatistics, School of Public Health, & The Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100083, China
| |
Collapse
|
283
|
Villacampa G, Tolosa P, Salvador F, Sánchez-Bayona R, Villanueva L, Dienstmann R, Ciruelos E, Pascual T. Addition of immune checkpoint inhibitors to chemotherapy versus chemotherapy alone in first-line metastatic triple-negative breast cancer: a systematic review and meta-analysis. Cancer Treat Rev 2022; 104:102352. [DOI: 10.1016/j.ctrv.2022.102352] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/31/2022]
|
284
|
Hao KJ, Jia X, Dai WT, Huo ZM, Zhang HQ, Liu JW, Wang XB. Mapping Intellectual Structures and Research Hotspots of Triple Negative Breast Cancer: A Bibliometric Analysis. Front Oncol 2022; 11:689553. [PMID: 35047380 PMCID: PMC8763010 DOI: 10.3389/fonc.2021.689553] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/30/2021] [Indexed: 01/12/2023] Open
Abstract
Background Triple negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype with a poor prognosis due to its extremely aggressive nature and lack of effective treatment options. This study aims to summarize the current hotspots of TNBC research and evaluate the TNBC research trends, both qualitatively and quantitatively. Methods Scientific publications of TNBC-related studies from January 1, 2010 to October 17, 2020 were obtained from the Web of Science database. The BICOMB software was used to obtain the high-frequency keywords layout. The gCLUTO was used to produce a biclustering analysis on the binary matrix of word-paper. The co-occurrence and collaboration analysis between authors, countries, institutions, and keywords were performed by VOSviewer software. Keyword burst detection was performed by CiteSpace. Results A total of 12,429 articles related to TNBC were identified. During 2010-2020, the most productive country/region and institution in TNBC field was the USA and The University of Texas MD Anderson Cancer Center, respectively. Cancer Research, Journal of Clinical Oncology, and Annals of Oncology were the first three periodicals with maximum publications in TNBC research. Eight research hotspots of TNBC were identified by co-word analysis. In the core hotspots, research on neoadjuvant chemotherapy, paclitaxel therapy, and molecular typing of TNBC is relatively mature. Research on immunotherapy and PARP inhibitor for TNBC is not yet mature but is the current focus of this field. Burst detection of keywords showed that studies on TNBC proteins and receptors, immunotherapy, target, and tumor cell migration showed bursts in recent three years. Conclusion The current study revealed that TNBC studies are growing. Attention should be paid to the latest hotspots, such as immunotherapy, PARP inhibitors, target, and TNBC proteins and receptors.
Collapse
Affiliation(s)
- Kai-Jun Hao
- Department of Plastic Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiao Jia
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
| | - Wen-Ting Dai
- Department of Plastic Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Ze-Min Huo
- Department of Plastic Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Hua-Qiang Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing-Wei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiao-Bing Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
285
|
Abe S, Nagata H, Crosby EJ, Inoue Y, Kaneko K, Liu CX, Yang X, Wang T, Acharya CR, Agarwal P, Snyder J, Gwin W, Morse MA, Zhong P, Lyerly HK, Osada T. Combination of ultrasound-based mechanical disruption of tumor with immune checkpoint blockade modifies tumor microenvironment and augments systemic antitumor immunity. J Immunother Cancer 2022; 10:jitc-2021-003717. [PMID: 35039461 PMCID: PMC8765068 DOI: 10.1136/jitc-2021-003717] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 02/02/2023] Open
Abstract
Background Despite multimodal adjuvant management with radiotherapy, chemotherapy and hormonal therapies, most surgically resected primary breast cancers relapse or metastasize. A potential solution to late and distant recurrence is to augment systemic antitumor immunity, in part by appropriately presenting tumor antigens, but also by modulating the immunosuppressive tumor microenvironment (TME). We previously validated this concept in models of murine carcinoma treated with a novel predominately microcavitating version of high-intensity focused ultrasound (HIFU), mechanical high-intensity focused ultrasound (M-HIFU). Here we elucidated the mechanisms of enhanced antitumor immunity by M-HIFU over conventional thermal high-intensity focused ultrasound (T-HIFU) and investigated the potential of the combinatorial strategy with an immune checkpoint inhibitor, anti-PD-L1 antibody. Methods The antitumor efficacy of treatments was investigated in syngeneic murine breast cancer models using triple-negative (E0771) or human ErbB-2 (HER2) expressing (MM3MG-HER2) tumors in C57BL/6 or BALB/c mice, respectively. Induction of systemic antitumor immunity by the treatments was tested using bilateral tumor implantation models. Flow cytometry, immunohistochemistry, and single-cell RNA sequencing were performed to elucidate detailed effects of HIFU treatments or combination treatment on TME, including the activation status of CD8 T cells and polarization of tumor-associated macrophages (TAMs). Results More potent systemic antitumor immunity and tumor growth suppression were induced by M-HIFU compared with T-HIFU. Molecular characterization of the TME after M-HIFU by single-cell RNA sequencing demonstrated repolarization of TAM to the immunostimulatory M1 subtype compared with TME post-T-HIFU. Concurrent anti-PD-L1 antibody administration or depletion of CD4+ T cells containing a population of regulatory T cells markedly increased T cell-mediated antitumor immunity and tumor growth suppression at distant, untreated tumor sites in M-HIFU treated mice compared with M-HIFU monotherapy. CD8 T and natural killer cells played major roles as effector cells in the combination treatment. Conclusions Physical disruption of the TME by M-HIFU repolarizes TAM, enhances T-cell infiltration, and, when combined with anti-PD-L1 antibody, mediates superior systemic antitumor immune responses and distant tumor growth suppression. These findings suggest M-HIFU combined with anti-PD-L1 may be useful in reducing late recurrence or metastasis when applied to primary tumors.
Collapse
Affiliation(s)
- Shinya Abe
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Nagata
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Erika J Crosby
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Yoshiyuki Inoue
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgery, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kensuke Kaneko
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Surgical Oncology, Faculty of Medicine, The University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Cong-Xiao Liu
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Xiao Yang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Tao Wang
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Chaitanya R Acharya
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Pankaj Agarwal
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Joshua Snyder
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - William Gwin
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Michael A Morse
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA.,Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Pei Zhong
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
286
|
Newton EE, Mueller LE, Treadwell SM, Morris CA, Machado HL. Molecular Targets of Triple-Negative Breast Cancer: Where Do We Stand? Cancers (Basel) 2022; 14:482. [PMID: 35158750 PMCID: PMC8833442 DOI: 10.3390/cancers14030482] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer. Due to its heterogeneity and lack of hormone receptor expression, this subtype is more likely to metastasize and resist treatment attempts than are other forms of breast cancer. Due to the absence of targetable receptors, chemotherapy and breast conserving surgery have been the predominant treatment options for patients. However, resistance to chemotherapy and local recurrence of the tumors is frequent. Emerging immunotherapies have begun to change treatment plans for patients diagnosed with TNBC. In this review, we discuss the various immune pathways identified in TNBC and the role they play as targets for new potential treatment choices. Various therapeutic options that inhibit key pathways in cellular growth cycles, DNA repair mechanisms, epithelial mesenchymal transition, and immunosuppression have been shown to improve survival in patients with this disease. With promising results thus far, continued studies of immunotherapy and neoadjuvant therapy options for TNBC are likely to alter the treatment course for these diagnoses in the future.
Collapse
Affiliation(s)
- Emma E. Newton
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (E.E.N.); (L.E.M.); (S.M.T.)
| | - Lauren E. Mueller
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (E.E.N.); (L.E.M.); (S.M.T.)
| | - Scout M. Treadwell
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (E.E.N.); (L.E.M.); (S.M.T.)
| | - Cindy A. Morris
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Heather L. Machado
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (E.E.N.); (L.E.M.); (S.M.T.)
- Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA 70112, USA
| |
Collapse
|
287
|
St-Denis-Bissonnette F, Khoury R, Mediratta K, El-Sahli S, Wang L, Lavoie JR. Applications of Extracellular Vesicles in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:451. [PMID: 35053616 PMCID: PMC8773485 DOI: 10.3390/cancers14020451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and refractory subtype of breast cancer, often occurring in younger patients with poor clinical prognosis. Given the current lack of specific targets for effective intervention, the development of better treatment strategies remains an unmet medical need. Over the last decade, the field of extracellular vesicles (EVs) has grown tremendously, offering immense potential for clinical diagnosis/prognosis and therapeutic applications. While TNBC-EVs have been shown to play an important role in tumorigenesis, chemoresistance and metastasis, they could be repurposed as potential biomarkers for TNBC diagnosis and prognosis. Furthermore, EVs from various cell types can be utilized as nanoscale drug delivery systems (NDDS) for TNBC treatment. Remarkably, EVs generated from specific immune cell subsets have been shown to delay solid tumour growth and reduce tumour burden, suggesting a new immunotherapy approach for TNBC. Intrinsically, EVs can cross the blood-brain barrier (BBB), which holds great potential to treat the brain metastases diagnosed in one third of TNBC patients that remains a substantial clinical challenge. In this review, we present the most recent applications of EVs in TNBC as diagnostic/prognostic biomarkers, nanoscale drug delivery systems and immunotherapeutic agents, as well as discuss the associated challenges and future directions of EVs in cancer immunotherapy.
Collapse
Affiliation(s)
- Frederic St-Denis-Bissonnette
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Rachil Khoury
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Jessie R. Lavoie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| |
Collapse
|
288
|
Abstract
The widespread adoption of immunotherapy has revolutionized the treatment of various cancer types, including metastatic triple-negative breast cancer (TNBC), which has long been associated with poor prognostic outcomes. In particular, immune checkpoint inhibitors (ICIs) that target and inhibit programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1), have shown promising results in the treatment of patients with metastatic TNBC. However, while manipulating the immune system to induce antitumor response, ICIs can also lead to a unique set of immune-related adverse events (IRAEs), which differ from standard chemotherapy toxicities due to their immune-based origin. These toxicities require highly specific management, including guidance from multidisciplinary specialists. The primary treatment strategy against IRAEs is systemic corticosteroid use, but additional treatment approaches may also involve supportive care, additional immunosuppression, and concurrent treatment delay or discontinuation. Given the rising prevalence of ICI therapy, it is essential to educate clinicians on the presentation and management of these potentially life-threatening events so that they are identified early and treated appropriately. Using data from recent clinical trials, this review will focus on known IRAEs, particularly those seen in patients with breast cancer, and will summarize their prevalence, severity, and outcomes. We will discuss optimal strategies for early recognition and management, as well as approaches toward cautious retreatment following resolution of IRAEs.
Collapse
|
289
|
Schlauch D, Fu X, Jones SF, Burris HA, Spigel DR, Reeves J, McKenzie AJ. Tumor-Specific and Tumor-Agnostic Molecular Signatures Associated With Response to Immune Checkpoint Inhibitors. JCO Precis Oncol 2022; 5:1625-1638. [PMID: 34994650 DOI: 10.1200/po.21.00008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Next-generation sequencing (NGS) testing is being incorporated into routine standard of care for patients with cancer. Immune checkpoint inhibitors (CPIs) are approved for use in both tumor-specific and tumor-agnostic indications. We sought to determine tumor type-specific or tumor-agnostic correlations between mutations detected by NGS and response to CPIs. MATERIALS AND METHODS A retrospective analysis of 26,004 patient records with NGS data available was conducted. Time to treatment failure and overall survival analyses were performed. Hazard ratios and associated statistics were computed in the R programming language. The study was considered exempt from internal review board review and data were considered nonhuman subjects. RESULTS Response to CPIs varied between tumor types with melanoma and lung cancer performing relatively better on CPIs than other tumor types. Within tumor types, response to CPIs was stratified by mutations in specific genes. Tumor-agnostic markers including high tumor mutation burden and microsatellite instability-high were also associated with longer time to treatment failure on CPIs. Importantly, within the high tumor mutation burden and microsatellite instability-high groups, mutations in individual genes correlate with response to CPIs. CONCLUSION The results from commercial NGS panels may be used to stratify patients for response to CPIs. In tumors where CPIs show relatively low efficacy, there may be distinct patient populations-based on gene mutation status-that are predicted to have better response to CPIs. Likewise, there may be distinct patient populations who do relatively worse on CPIs within tumor types known to respond well to CPIs.
Collapse
Affiliation(s)
- Daniel Schlauch
- Sarah Cannon Research Institute, Nashville, TN.,Genospace, Boston, MA
| | - Xiaotong Fu
- Sarah Cannon Research Institute, Nashville, TN.,Genospace, Boston, MA
| | | | - Howard A Burris
- Sarah Cannon Research Institute, Nashville, TN.,Tennessee Oncology, PLLC, Nashville, TN
| | - David R Spigel
- Sarah Cannon Research Institute, Nashville, TN.,Tennessee Oncology, PLLC, Nashville, TN
| | | | | |
Collapse
|
290
|
Chen F, Chen N, Gao Y, Jia L, Lyu Z, Cui J. Clinical Progress of PD-1/L1 Inhibitors in Breast Cancer Immunotherapy. Front Oncol 2022; 11:724424. [PMID: 35070955 PMCID: PMC8770281 DOI: 10.3389/fonc.2021.724424] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is a major killer of women's health worldwide. While breast cancer is thought to have lower immunogenicity compared with other solid tumors, combination therapy is able to improve the immunogenicity of the tumor and sensitize breast cancer cells to immunotherapy. Immunotherapy represented by immune checkpoint inhibitors (ICIs) has been largely explored in the field of breast cancer, including both early and advanced disease. Immunotherapy for triple-negative breast cancer (TNBC) has been the most studied, and the PD-L1 inhibitor atezolizumab combined with nab-paclitaxel has been used in the first-line treatment of TNBC. Immunotherapeutic data for human epidermal growth factor receptor-positive and hormone receptor-positive breast cancer are also accumulating. This review summarizes the clinical trial data of ICIs or ICI-containing therapies in different types and stages of breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
291
|
DiMarco AV, Qin X, McKinney BJ, Garcia NMG, Van Alsten SC, Mendes EA, Force J, Hanks BA, Troester MA, Owzar K, Xie J, Alvarez JV. APOBEC Mutagenesis Inhibits Breast Cancer Growth through Induction of T cell-Mediated Antitumor Immune Responses. Cancer Immunol Res 2022; 10:70-86. [PMID: 34795033 PMCID: PMC8957077 DOI: 10.1158/2326-6066.cir-21-0146] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/23/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022]
Abstract
The APOBEC family of cytidine deaminases is one of the most common endogenous sources of mutations in human cancer. Genomic studies of tumors have found that APOBEC mutational signatures are enriched in the HER2 subtype of breast cancer and are associated with immunotherapy response in diverse cancer types. However, the direct consequences of APOBEC mutagenesis on the tumor immune microenvironment have not been thoroughly investigated. To address this, we developed syngeneic murine mammary tumor models with inducible expression of APOBEC3B. We found that APOBEC activity induced antitumor adaptive immune responses and CD4+ T cell-mediated, antigen-specific tumor growth inhibition. Although polyclonal APOBEC tumors had a moderate growth defect, clonal APOBEC tumors were almost completely rejected, suggesting that APOBEC-mediated genetic heterogeneity limits antitumor adaptive immune responses. Consistent with the observed immune infiltration in APOBEC tumors, APOBEC activity sensitized HER2-driven breast tumors to anti-CTLA-4 checkpoint inhibition and led to a complete response to combination anti-CTLA-4 and anti-HER2 therapy. In human breast cancers, the relationship between APOBEC mutagenesis and immunogenicity varied by breast cancer subtype and the frequency of subclonal mutations. This work provides a mechanistic basis for the sensitivity of APOBEC tumors to checkpoint inhibitors and suggests a rationale for using APOBEC mutational signatures and clonality as biomarkers predicting immunotherapy response in HER2-positive (HER2+) breast cancers.
Collapse
Affiliation(s)
- Ashley V DiMarco
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Xiaodi Qin
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Brock J McKinney
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Nina Marie G Garcia
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Sarah C Van Alsten
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Elizabeth A Mendes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Jeremy Force
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Durham, North Carolina
| | - Brent A Hanks
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Durham, North Carolina
| | - Melissa A Troester
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Jichun Xie
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - James V Alvarez
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
292
|
Zhang L, Chen Y, Lv Y, Jiao S, Zhao W. OUP accepted manuscript. Oncologist 2022; 27:245-250. [PMID: 35380719 DOI: 10.1093/oncolo/oyac027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 12/28/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- Li Zhang
- Department of Oncology, PLA General Hospital, Beijing, People's Republic of China
| | - Yimeng Chen
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, People's Republic of China
| | - Yao Lv
- Department of Oncology, PLA General Hospital, Beijing, People's Republic of China
| | - Shunchang Jiao
- Department of Oncology, PLA General Hospital, Beijing, People's Republic of China
| | - Weihong Zhao
- Department of Oncology, PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
293
|
Evaluating mismatch repair status to screen clinical advanced breast carcinomas for immunotherapy: experience from a large academic institution. Clin Breast Cancer 2022; 22:e680-e684. [DOI: 10.1016/j.clbc.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022]
|
294
|
Affiliation(s)
- Ilana Schlam
- Division of Hematology and Oncology, Tufts Medical Center, Boston, MA, USA
| | - Margaret E Gatti-Mays
- Corresponding author: Margaret E. Gatti-Mays, MD, MPH, 1800 Cannon Drive, 1300A Lincoln Tower, Columbus, OH 43210, USA. Tel: +1 614 366 2698;
| |
Collapse
|
295
|
Chan WL, Marinho J, Chavarri-Guerra Y, Hincapie-Echeverri J, Velasco RN, Akagunduz B, Roy M, Kwong WTG, Wu WF, Battisti NML, Soto-Perez-de-Celis E. Systemic treatment for triple negative breast cancer in older patients: A Young International Society of Geriatric Oncology Review Paper. J Geriatr Oncol 2022; 13:563-571. [DOI: 10.1016/j.jgo.2022.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/04/2022] [Indexed: 12/27/2022]
|
296
|
Chera A, Stancu AL, Bucur O. Thyroid-related adverse events induced by immune checkpoint inhibitors. Front Endocrinol (Lausanne) 2022; 13:1010279. [PMID: 36204105 PMCID: PMC9530140 DOI: 10.3389/fendo.2022.1010279] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Immune checkpoint inhibitors, namely anti-CTLA-4, anti-PD-1 and anti-PD-L1 monoclonal antibodies, have emerged in the last decade as a novel form of cancer treatment, promoting increased survival in patients. As they tamper with the immune response in order to destroy malignant cells, a new type of adverse reactions has emerged, known as immune-related adverse events (irAEs), which frequently target the endocrine system, especially the thyroid and hypophysis. Thyroid irAEs include hyperthyroidism, thyrotoxicosis, hypothyroidism and a possibly life-threatening condition known as the "thyroid storm". Early prediction of occurrence and detection of the thyroid irAEs should be a priority for the clinician, in order to avoid critical situations. Moreover, they are recently considered both a prognostic marker and a means of overseeing treatment response, since they indicate an efficient activation of the immune system. Therefore, a multidisciplinary approach including both oncologists and endocrinologists is recommended when immune checkpoint inhibitors are used in the clinic.
Collapse
Affiliation(s)
- Alexandra Chera
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andreea Lucia Stancu
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Octavian Bucur
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Viron Molecular Medicine Institute, Boston, MA, United States
- *Correspondence: Octavian Bucur, ;;
| |
Collapse
|
297
|
Yang F, Wang JF, Wang Y, Liu B, Molina JR. Comparative Analysis of Predictive Biomarkers for PD-1/PD-L1 Inhibitors in Cancers: Developments and Challenges. Cancers (Basel) 2021; 14:cancers14010109. [PMID: 35008273 PMCID: PMC8750062 DOI: 10.3390/cancers14010109] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The development of immune checkpoint inhibitors (ICIs) has greatly changed the treatment landscape of multiple malignancies. However, the wide administration of ICIs is mainly obstructed by the low response rate and several life-threatening adverse events. Thus, there is an urgent need to identify sets of biomarkers to predict which patients will respond to ICIs. In this review, we discuss the recently investigated molecular and clinical determinants of ICI response, from the aspects of tumor features, clinical features, as well as tumor microenvironment. Abstract Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) have dramatically changed the landscape of cancer therapy. Both remarkable and durable responses have been observed in patients with melanoma, non-small-cell lung cancer (NSCLC), and other malignancies. However, the PD-1/PD-L1 blockade has demonstrated meaningful clinical responses and benefits in only a subset of patients. In addition, several severe and life-threatening adverse events were observed in these patients. Therefore, the identification of predictive biomarkers is urgently needed to select patients who are more likely to benefit from ICI therapy. PD-L1 expression level is the most commonly used biomarker in clinical practice for PD-1/PD-L1 inhibitors. However, negative PD-L1 expression cannot reliably exclude a response to a PD-1/PD-L1 blockade. Other factors, such as tumor microenvironment and other tumor genomic signatures, appear to impact the response to ICIs. In this review, we examine emerging data for novel biomarkers that may have a predictive value for optimizing the benefit from anti-PD-1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Fang Yang
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China;
| | | | - Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Baorui Liu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China;
- Correspondence: (B.L.); (J.R.M.)
| | - Julian R. Molina
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: (B.L.); (J.R.M.)
| |
Collapse
|
298
|
Santisteban M, Solans BP, Hato L, Urrizola A, Mejías LD, Salgado E, Sánchez-Bayona R, Toledo E, Rodríguez-Spiteri N, Olartecoechea B, Idoate MA, López-Díaz de Cerio A, Inogés S. Final results regarding the addition of dendritic cell vaccines to neoadjuvant chemotherapy in early HER2-negative breast cancer patients: clinical and translational analysis. Ther Adv Med Oncol 2021; 13:17588359211064653. [PMID: 34987618 PMCID: PMC8721381 DOI: 10.1177/17588359211064653] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Primary breast cancer (BC) has shown a higher immune infiltration than the metastatic disease, justifying the optimal scenario for immunotherapy. Recently, neoadjuvant chemotherapy (NAC) combined with immune checkpoint inhibitors has demonstrated a gain in pathological complete responses (tpCR) in patients with BC. The aim of our study is to evaluate the safety, feasibility, and efficacy of the addition of dendritic cell vaccines (DCV) to NAC in HER2-negative BC patients. METHODS Thirty-nine patients with early BC received DCV together with NAC conforming the vaccinated group (VG) and compared with 44 patients as the control group (CG). All patients received anthracyclines and taxanes-based NAC (ddECx4→Dx4) followed by surgery ± radiotherapy ± hormonotherapy. RESULTS The tpCR rate was 28.9% in the VG and 9.09% in the CG (p = 0.03). Pathological CR in the triple negative (TN) BC were 50.0% versus 30.7% (p = 0.25), 16.6% versus 0% in luminal B (p = 0.15), and none among luminal A patients in VG versus CG, respectively. Impact of DCV was significantly higher in the programmed cell death ligand 1 (PD-L1) negative population (p < 0.001). PD-L1 expression was increased in patients with residual disease in the VG as compared with the CG (p < 0.01). No grade ⩾3 vaccine-related adverse events occurred. With a median follow-up of 8 years, no changes were seen in event-free survival or overall survival. Phenotypic changes post DCV in peripheral blood were observed in myeloid-derived suppressor cells (MDSC), NK, and T cells. Increase in blood cell proliferation and interferon (IFN)-γ production was detected in 69% and 74% in the VG, respectively. Humoral response was also found. Clonality changes in TCR-β repertoire were detected in 67% of the patients with a drop in diversity index after treatment. CONCLUSION The combination of DCV plus NAC is safe and increases tpCR, with a significant benefit among PD-L1-negative tumors. DCV modify tumor milieu and perform cellular and humoral responses in peripheral blood with no impact in outcome. TRIAL REGISTRATION ClinicalTrials.gov number: NCT01431196. EudraCT 2009-017402-36.
Collapse
Affiliation(s)
- Marta Santisteban
- Department of Medical Oncology, Clínica Universidad de Navarra, Avda. Pío XII 36, 31008 Pamplona, Spain
- Breast Cancer Unit, Clínica Universidad de Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Belén Pérez Solans
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Pharmacometrics and Systems Pharmacology, Universidad de Navarra, Pamplona, Spain
| | - Laura Hato
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Urrizola
- Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Luis Daniel Mejías
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Esteban Salgado
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Medical Oncology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | | | - Estefanía Toledo
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Department of Preventive Medicine and Public Health, Universidad de Navarra, Pamplona, Spain
| | | | | | | | - Ascensión López-Díaz de Cerio
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Cell Therapy Unit, Clínica Universidad de Navarra, Pamplona, Spain
- Clínica Universidad de Navarra, Universidad de Navarra, Complejo Hospitalario de Navarra and IdisNA, Pamplona, Spain
| | - Susana Inogés
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Cell Therapy Unit, Clínica Universidad de Navarra, Pamplona, Spain
- Clínica Universidad de Navarra, Universidad de Navarra, Complejo Hospitalario de Navarra and IdisNA, Pamplona, Spain
| |
Collapse
|
299
|
Dias CJ, Helguero L, Faustino MAF. Current Photoactive Molecules for Targeted Therapy of Triple-Negative Breast Cancer. Molecules 2021; 26:7654. [PMID: 34946732 PMCID: PMC8709347 DOI: 10.3390/molecules26247654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 01/10/2023] Open
Abstract
Cancer is the second leading cause of death worldwide; therefore, there is an urgent need to find safe and effective therapies. Triple-negative breast cancer (TNBC) is diagnosed in ca. 15-20% of BC and is extremely aggressive resulting in reduced survival rate, which is mainly due to the low therapeutic efficacy of available treatments. Photodynamic therapy (PDT) is an interesting therapeutic approach in the treatment of cancer; the photosensitizers with good absorption in the therapeutic window, combined with their specific targeting of cancer cells, have received particular interest. This review aims to revisit the latest developments on chlorin-based photoactive molecules for targeted therapy in TNBC. Photodynamic therapy, alone or combined with other therapies (such as chemotherapy or photothermal therapy), has potential to be a safe and a promising approach against TNBC.
Collapse
Affiliation(s)
- Cristina J. Dias
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Luisa Helguero
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
| | | |
Collapse
|
300
|
Moore J, Ma L, Lazar AA, Barcellos-Hoff MH. Mammary tumor-derived transplants as breast cancer models to evaluate tumor-immune interactions and therapeutic responses. Cancer Res 2021; 82:365-376. [PMID: 34903599 DOI: 10.1158/0008-5472.can-21-0253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/25/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022]
Abstract
In breast cancer, the type and distribution of infiltrating immune cells are associated with clinical outcome. Moreover, infiltrated cancers with abundant tumor infiltrating lymphocytes (TIL) are more likely to respond to immunotherapy, while those in which CD8+ T cells are completely absent (deserts) or excluded are less likely to respond. Detailed understanding of this biology is limited by a lack of preclinical breast cancer models that recapitulate TIL distributions and their associated biology. Here we established mammary tumor-derived transplants (mTDT) from 12 Trp53 null mammary tumors in syngeneic BALB/cJ mice and examined the stability of their growth rate, TIL distribution, and transcriptomic profiles. All mTDT were estrogen receptor negative. Half of the parental tumors were classified as infiltrated, and the rest were divided between excluded and desert phenotypes. After two orthotopic passages, most (70%) mTDT from infiltrated parents recapitulated this pattern, whereas the desert or excluded parental patterns were maintained in about half of daughter mTDT. Approximately 30% of mTDT gave rise to lung or liver metastases, but metastasis was not associated with a TIL phenotype. Unsupervised transcriptomic analysis clustered mTDT according to their TIL spatial patterns. Infiltrated mTDT transplanted subcutaneously and orthotopically were resistant to anti-PD-L1. Profiling implicated prolonged antigen stimulation and loss of effector function of lymphocytes rather than T cell exhaustion in the lack of response of infiltrated mTDT to checkpoint blockade. In summary, the molecular diversity and immune complexity of mTDT will facilitate the dissection of mechanisms of breast cancer response to immunotherapies.
Collapse
Affiliation(s)
- Jade Moore
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Lin Ma
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Ann A Lazar
- Department of Oral Epidemiology, School of Dentistry, University of California San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California San Francisco School of Medicine, San Francisco, California
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.
| |
Collapse
|