151
|
Tumor cells suppress radiation-induced immunity by hijacking caspase 9 signaling. Nat Immunol 2020; 21:546-554. [PMID: 32231300 DOI: 10.1038/s41590-020-0641-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 02/21/2020] [Indexed: 12/19/2022]
Abstract
High-dose radiation activates caspases in tumor cells to produce abundant DNA fragments for DNA sensing in antigen-presenting cells, but the intrinsic DNA sensing in tumor cells after radiation is rather limited. Here we demonstrate that irradiated tumor cells hijack caspase 9 signaling to suppress intrinsic DNA sensing. Instead of apoptotic genomic DNA, tumor-derived mitochondrial DNA triggers intrinsic DNA sensing. Specifically, loss of mitochondrial DNA sensing in Casp9-/- tumors abolishes the enhanced therapeutic effect of radiation. We demonstrated that combining emricasan, a pan-caspase inhibitor, with radiation generates synergistic therapeutic effects. Moreover, loss of CASP9 signaling in tumor cells led to adaptive resistance by upregulating programmed death-ligand 1 (PD-L1) and resulted in tumor relapse. Additional anti-PD-L1 blockade can further overcome this acquired immune resistance. Therefore, combining radiation with a caspase inhibitor and anti-PD-L1 can effectively control tumors by sequentially blocking both intrinsic and extrinsic inhibitory signaling.
Collapse
|
152
|
Costa RLB, Czerniecki BJ. Clinical development of immunotherapies for HER2 + breast cancer: a review of HER2-directed monoclonal antibodies and beyond. NPJ Breast Cancer 2020; 6:10. [PMID: 32195333 PMCID: PMC7067811 DOI: 10.1038/s41523-020-0153-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer accounts for ~25% of breast cancer cases. Monoclonal antibodies (mAbs) against HER2 have led to unparalleled clinical benefit for a subset of patients with HER2+ breast cancer. In this narrative review, we summarize advances in the understanding of immune system interactions, examine clinical developments, and suggest rationales for future investigation of immunotherapies for HER2+ breast cancer. Complex interactions have been found between different branches of the immune system, HER2+ breast cancer, and targeted treatments (approved and under investigation). A new wave of immunotherapies, such as novel HER2-directed mAbs, antibody drug conjugates, vaccines, and adoptive T-cell therapies, are being studied in a broad population of patients with HER2-expressing tumors. The development of immunotherapies for HER2+ breast cancer represents an evolving field that should take into account interactions between different components of the immune system.
Collapse
Affiliation(s)
- Ricardo L B Costa
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Brian J Czerniecki
- Departments of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| |
Collapse
|
153
|
Sow HS, Benonisson H, Brouwers C, Linssen MM, Camps M, Breukel C, Claassens J, van Hall T, Ossendorp F, Fransen MF, Verbeek JS. Immunogenicity of rat-neu + mouse mammary tumours determines the T cell-dependent therapeutic efficacy of anti-neu monoclonal antibody treatment. Sci Rep 2020; 10:3933. [PMID: 32127568 PMCID: PMC7054273 DOI: 10.1038/s41598-020-60893-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 02/14/2020] [Indexed: 11/09/2022] Open
Abstract
The use of Trastuzumab (Herceptin), a monoclonal antibody (mAb) targeting HER2/neu, results in an increased median survival in Her2+ breast cancer patients. The tumour mutational burden and the presence of tumour infiltrating lymphocytes (TILs) clearly correlate with response to trastuzumab. Here, we investigated if the immunogenicity of the transplantable rat-neu+ tumour cell line (TUBO) derived from a BALB/c-NeuT primary tumour is associated with the response to anti-neu mAb therapy. We compared the TUBO tumour outgrowth and tumour infiltrating T cells in isogenic (BALB/c-NeuT) and non-isogenic (WT BALB/c) recipient mice. Furthermore, therapeutic efficacy of anti-neu mAb and the contribution of T cells were examined in both mouse strains. The outgrowth of untreated tumours was significantly better in BALB/c-NeuT than WT BALB/c mice. Moreover, tumour infiltrating T cells were more abundantly present in WT BALB/c than BALB/c-NeuT mice, showing that the TUBO tumour was more immunogenic in WT BALB/c mice. In TUBO tumour bearing WT BALB/c mice, anti-neu mAb therapy resulted in an increase of tumour infiltrating T cells and long-term survival. When T cells were depleted, this strong anti-tumour effect was reduced to an outgrowth delay. In contrast, in TUBO tumour bearing BALB/c-NeuT mice, treatment with anti-neu mAb resulted only in tumour outgrowth delay, both in the presence and absence of T cells. We concluded that in immunogenic tumours the response to anti-neu mAb therapy is enhanced by additional T cell involvement compared to the response to anti-neu mAb in non-immunogenic tumours.
Collapse
Affiliation(s)
- Heng Sheng Sow
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Conny Brouwers
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Margot M Linssen
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marcel Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Cor Breukel
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jill Claassens
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Pulmonary Diseases, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Centre, Leiden, The Netherlands.
| |
Collapse
|
154
|
Sharma P, Connolly RM, Roussos Torres ET, Thompson A. Best Foot Forward: Neoadjuvant Systemic Therapy as Standard of Care in Triple-Negative and HER2-Positive Breast Cancer. Am Soc Clin Oncol Educ Book 2020; 40:1-16. [PMID: 32315235 DOI: 10.1200/edbk_281381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Neoadjuvant systemic treatment of early-stage breast cancer has been used to improve resectability and reduce the extent of breast and axillary surgery. More recently, several other merits of neoadjuvant systemic treatment have emerged, including the ability to tailor clinically available adjuvant systemic therapy options based on pathologic response and to serve as a platform for early assessment of novel agents and response biomarkers and as an avenue for treatment optimization investigations (local and systemic therapy escalation and de-escalation trials guided by pathologic response). Attainment of a pathologic complete response (pCR) is associated with excellent long-term outcomes; conversely, the presence of residual disease is associated with a high risk of recurrence for patients with HER2-positive breast cancer and triple-negative breast cancer (TNBC). Treatment strategies in early-stage HER2-positive breast cancer include regimens incorporating trastuzumab, pertuzumab, ado-trastuzumab emtansine, and neratinib, resulting in high pCR rates and overall excellent long-term outcomes. Currently available cytotoxic regimens yield pCR for 35% to 55% of patients with TNBC, and immune checkpoint inhibition is showing early promise for this subtype. New drug and predictive biomarker evaluations in the neoadjuvant setting aim to develop optimal treatment strategies for the individual patient, with the ultimate goal of maximizing efficacy and minimizing toxicity. Research efforts involving novel agents are being undertaken to address the high risk of recurrence for patients with residual disease. Omission of breast surgery following neoadjuvant chemotherapy requires further development of imaging and biopsy techniques to accurately assess the extent of residual disease before clinical application.
Collapse
Affiliation(s)
- Priyanka Sharma
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Westwood, KS
| | | | | | - Alastair Thompson
- Department of Surgery, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
155
|
Zhang Z, Xie T, Zhang X, Qi C, Shen L, Peng Z. Immune checkpoint inhibitors for treatment of advanced gastric or gastroesophageal junction cancer: Current evidence and future perspectives. Chin J Cancer Res 2020; 32:287-302. [PMID: 32694895 PMCID: PMC7369180 DOI: 10.21147/j.issn.1000-9604.2020.03.02] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite the application of conventional therapies, the prognosis of advanced gastric cancer (GC) or gastroesophageal junction cancer (GEJC) is still poor. In recent years, immune checkpoint inhibitors (ICIs) have reshaped the paradigm of cancer therapy. Emerging evidence support the feasibility of programmed cell death-1 (PD-1) and its ligand (PD-L1) inhibition in chemo-refractory GC/GEJC. Nivolumab and pembrolizumab have initially been approved in Japan and United States, respectively for the third-line treatment of progressive GC or GEJC. In March 2020, nivolumab has also been licensed in China for treating advanced GC/GEJC who received ≥2 lines of systemic therapies. Current studies are moving forward to the first-line application or focusing on combination strategies, though data are insufficient and disputable. In this review, we summarize the recently reported and ongoing clinical trials in ICIs for advanced GC/GEJC. Molecular characteristics and clinical implications of different tumor subtypes are also reviewed. We further discuss the safety profile and biomarkers for predicting the response of ICIs, which has guiding values in clinical practice.
Collapse
Affiliation(s)
- Zhening Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Tong Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaotian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Changsong Qi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Peng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
156
|
Jang BS, Han W, Kim IA. Tumor mutation burden, immune checkpoint crosstalk and radiosensitivity in single-cell RNA sequencing data of breast cancer. Radiother Oncol 2020; 142:202-209. [DOI: 10.1016/j.radonc.2019.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/23/2019] [Accepted: 11/04/2019] [Indexed: 02/04/2023]
|
157
|
Scott E, Elliott DJ, Munkley J. Tumour associated glycans: A route to boost immunotherapy? Clin Chim Acta 2019; 502:167-173. [PMID: 31870793 DOI: 10.1016/j.cca.2019.12.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
While the development of immunotherapies for cancer treatment offer significant promise across several cancers, still only a small subset of patients respond to immune based monotherapies. As such, attention has turned to the development of combination therapies. These use conventional cancer treatments such as chemotherapy to sensitise tumours to immunotherapy. Here, we summarise key research, highlighting the exciting potential of tumour associated glycans as therapeutic targets to sensitise tumours to immunotherapy. When cells undergo carcinogenesis they reprogram their glyco-code. Several cancer associated glycans have been identified, and therapies targeting them are under development. Proteins containing carbohydrate binding domains (lectins) are expressed by many immune cell subtypes, and upon glycan binding, transduce immune modulatory signals that regulate the tumour immune microenvironment.
Collapse
Affiliation(s)
- Emma Scott
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK.
| | - David J Elliott
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
| | - Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
| |
Collapse
|
158
|
Tsao LC, Crosby EJ, Trotter TN, Agarwal P, Hwang BJ, Acharya C, Shuptrine CW, Wang T, Wei J, Yang X, Lei G, Liu CX, Rabiola CA, Chodosh LA, Muller WJ, Lyerly HK, Hartman ZC. CD47 blockade augmentation of trastuzumab antitumor efficacy dependent on antibody-dependent cellular phagocytosis. JCI Insight 2019; 4:131882. [PMID: 31689243 PMCID: PMC6975273 DOI: 10.1172/jci.insight.131882] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022] Open
Abstract
The HER2-specific monoclonal antibody (mAb), trastuzumab, has been the mainstay of therapy for HER2+ breast cancer (BC) for approximately 20 years. However, its therapeutic mechanism of action (MOA) remains unclear, with antitumor responses to trastuzumab remaining heterogeneous and metastatic HER2+ BC remaining incurable. Consequently, understanding its MOA could enable rational strategies to enhance its efficacy. Using both murine and human versions of trastuzumab, we found its antitumor activity dependent on Fcγ receptor stimulation of tumor-associated macrophages (TAMs) and antibody-dependent cellular phagocytosis (ADCP), but not cellular cytotoxicity (ADCC). Trastuzumab also stimulated TAM activation and expansion, but did not require adaptive immunity, natural killer cells, and/or neutrophils. Moreover, inhibition of the innate immune ADCP checkpoint, CD47, significantly enhanced trastuzumab-mediated ADCP and TAM expansion and activation, resulting in the emergence of a unique hyperphagocytic macrophage population, improved antitumor responses, and prolonged survival. In addition, we found that tumor-associated CD47 expression was inversely associated with survival in HER2+ BC patients and that human HER2+ BC xenografts treated with trastuzumab plus CD47 inhibition underwent complete tumor regression. Collectively, our study identifies trastuzumab-mediated ADCP as an important antitumor MOA that may be clinically enabled by CD47 blockade to augment therapeutic efficacy.
Collapse
Affiliation(s)
- Li-Chung Tsao
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Erika J. Crosby
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | | | - Pankaj Agarwal
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Bin-Jin Hwang
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | | | | | - Tao Wang
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Junping Wei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Xiao Yang
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | | | - Lewis A. Chodosh
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William J. Muller
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Immunology, and
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Zachary C. Hartman
- Department of Surgery, Duke University, Durham, North Carolina, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
159
|
Targeting innate sensing in the tumor microenvironment to improve immunotherapy. Cell Mol Immunol 2019; 17:13-26. [PMID: 31844141 DOI: 10.1038/s41423-019-0341-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/24/2019] [Indexed: 12/14/2022] Open
Abstract
The innate immune sensing pathways play critical roles in the defense against pathogen infection, but their roles in cancer immunosurveillance and cancer therapies are less defined. We propose that defective innate immune sensing inside the tumor microenvironment might limit T-cell responses to immunotherapy. A recent mechanistic understanding of conventional therapies revealed that both innate immune sensing and T-cell responses are essential for optimal antitumor efficacy. T-cell-based immunotherapy, particularly immune checkpoint blockade, has achieved great success in reactivating antitumor immune responses to lead to tumor regression, but only in a small fraction of patients. Therefore, incorporating conventional therapy that can increase innate sensing and immunotherapy should lead to promising strategies for cancer patients. Here, we review the innate sensing pathways related to cancer initiation/progression and therapies, summarize the recent key findings in innate immune sensing related to conventional therapies, evaluate current combination strategies, and highlight the potential issues of combinational therapies in terms of antitumor efficacy and toxicities.
Collapse
|
160
|
Gastroesophageal cancer: Navigating the immune and genetic terrain to improve clinical outcomes. Cancer Treat Rev 2019; 84:101950. [PMID: 31918022 DOI: 10.1016/j.ctrv.2019.101950] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023]
Abstract
Recent advances in our understanding of the molecular biology of gastric and oesophageal cancers have shown that gastroesophageal adenocarcinoma should be considered as one disease spectrum. Clinical management of these cancers is challenging, with poor outcomes in both early and late disease settings. Certain molecular subsets of gastroesophageal adenocarcinoma demonstrate features that suggest immunotherapy could be an effective treatment. Immunogenetic markers, including mismatch repair deficiency, PD-L1 status and tumour infiltrating lymphocytes influence overall prognosis. They may also determine the response to adjuvant and neoadjuvant conventional chemotherapy. Initial results from immunotherapy trials for gastroesophageal cancer have however been mixed, with poor overall responses in the first- and second-line settings. This review aims to discuss how better understanding of these immune and genetic interactions may lead to better selection of patients for conventional and immune based therapies, and therefore improve patient outcomes. We also discuss the challenges in implementing this new understanding in routine practice, and the current limitations of immune based treatments for gastroesophageal cancer.
Collapse
|
161
|
Chun BM, Page DB, McArthur HL. Combination Immunotherapy Strategies in Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00333-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
We summarize combination immunotherapy strategies for the treatment of breast cancer, with a focus on metastatic disease. First, a general overview of combination approaches is presented according to breast cancer subtype. Second, additional review of promising combination approaches is presented.
Recent Findings
Combination strategies utilizing chemotherapy or radiotherapy with immune checkpoint inhibition are being evaluated across multiple phase III trials. Dual immunotherapy strategies, such as dual immune checkpoint inhibition or combined co-stimulation/co-inhibition, have supportive preclinical evidence and are under early clinical investigation. Modulation of the immune microenvironment via cytokines and vaccination strategies, as well as locally focused treatments to enhance antigenic responses, are active areas of research.
Summary
Pre-clinical and translational research sheds new light on numerous ways the immune system may be modulated to fight against cancer. We describe current and emerging combination approaches which may improve patient outcomes in metastatic breast cancer.
Collapse
|
162
|
Li XY, Moesta AK, Xiao C, Nakamura K, Casey M, Zhang H, Madore J, Lepletier A, Aguilera AR, Sundarrajan A, Jacoberger-Foissac C, Wong C, Dela Cruz T, Welch M, Lerner AG, Spatola BN, Soros VB, Corbin J, Anderson AC, Effern M, Hölzel M, Robson SC, Johnston RL, Waddell N, Smith C, Bald T, Geetha N, Beers C, Teng MWL, Smyth MJ. Targeting CD39 in Cancer Reveals an Extracellular ATP- and Inflammasome-Driven Tumor Immunity. Cancer Discov 2019; 9:1754-1773. [PMID: 31699796 DOI: 10.1158/2159-8290.cd-19-0541] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/04/2019] [Accepted: 10/01/2019] [Indexed: 01/24/2023]
Abstract
We explored the mechanism of action of CD39 antibodies that inhibit ectoenzyme CD39 conversion of extracellular ATP (eATP) to AMP and thus potentially augment eATP-P2-mediated proinflammatory responses. Using syngeneic and humanized tumor models, we contrast the potency and mechanism of anti-CD39 mAbs with other agents targeting the adenosinergic pathway. We demonstrate the critical importance of an eATP-P2X7-ASC-NALP3-inflammasome-IL18 pathway in the antitumor activity mediated by CD39 enzyme blockade, rather than simply reducing adenosine as mechanism of action. Efficacy of anti-CD39 activity was underpinned by CD39 and P2X7 coexpression on intratumor myeloid subsets, an early signature of macrophage depletion, and active IL18 release that facilitated the significant expansion of intratumor effector T cells. More importantly, anti-CD39 facilitated infiltration into T cell-poor tumors and rescued anti-PD-1 resistance. Anti-human CD39 enhanced human T-cell proliferation and Th1 cytokine production and suppressed human B-cell lymphoma in the context of autologous Epstein-Barr virus-specific T-cell transfer. SIGNIFICANCE: Overall, these data describe a potent and novel mechanism of action of antibodies that block mouse or human CD39, triggering an eATP-P2X7-inflammasome-IL18 axis that reduces intratumor macrophage number, enhances intratumor T-cell effector function, overcomes anti-PD-1 resistance, and potentially enhances the efficacy of adoptive T-cell transfer.This article is highlighted in the In This Issue feature, p. 1631.
Collapse
Affiliation(s)
- Xian-Yang Li
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Christos Xiao
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kyohei Nakamura
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mika Casey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Haiyan Zhang
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jason Madore
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ailin Lepletier
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Amelia Roman Aguilera
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ashmitha Sundarrajan
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Celia Jacoberger-Foissac
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | | | - Megan Welch
- Tizona Therapeutics, San Francisco, California
| | | | | | | | - John Corbin
- Tizona Therapeutics, San Francisco, California
| | - Ana C Anderson
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Maike Effern
- Unit of RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Michael Hölzel
- Unit of RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Simon C Robson
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Rebecca L Johnston
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Nicola Waddell
- Medical Genomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Corey Smith
- Translational and Human Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Tobias Bald
- Oncology and Cellular Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Nishamol Geetha
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.
| |
Collapse
|
163
|
Krasniqi E, Barchiesi G, Pizzuti L, Mazzotta M, Venuti A, Maugeri-Saccà M, Sanguineti G, Massimiani G, Sergi D, Carpano S, Marchetti P, Tomao S, Gamucci T, De Maria R, Tomao F, Natoli C, Tinari N, Ciliberto G, Barba M, Vici P. Immunotherapy in HER2-positive breast cancer: state of the art and future perspectives. J Hematol Oncol 2019; 12:111. [PMID: 31665051 PMCID: PMC6820969 DOI: 10.1186/s13045-019-0798-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/25/2019] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BC) is a complex disease with primary or acquired incurability characteristics in a significant part of patients. Immunotherapeutical agents represent an emerging option for breast cancer treatment, including the human epidermal growth factor 2 positive (HER2+) subtype. The immune system holds the ability to spontaneously implement a defensive response against HER2+ BC cells through complex mechanisms which can be exploited to modulate this response for obtaining a clinical benefit. Initial immune system modulating strategies consisted mostly in vaccine therapies, which are still being investigated and improved. However, the entrance of trastuzumab into the scenery of HER2+ BC treatment was the real game changing event, which embodied a dominant immune-mediated mechanism. More recently, the advent of the immune checkpoint inhibitors has caused a new paradigm shift for immuno-oncology, with promising initial results also for HER2+ BC. Breast cancer has been traditionally considered poorly immunogenic, being characterized by relatively low tumor mutation burden (TMB). Nevertheless, recent evidence has revealed high tumor infiltrating lymphocytes (TILs) and programmed cell death-ligand 1 (PD-L1) expression in a considerable proportion of HER2+ BC patients. This may translate into a higher potential to elicit anti-cancer response and, therefore, wider possibilities for the use and implementation of immunotherapy in this subset of BC patients. We are herein presenting and critically discussing the most representative evidence concerning immunotherapy in HER2+ BC cancer, both singularly and in combination with therapeutic agents acting throughout HER2-block, immune checkpoint inhibition and anti-cancer vaccines. The reader will be also provided with hints concerning potential future projection of the most promising immutherapeutic agents and approaches for the disease of interest.
Collapse
Affiliation(s)
- E Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - G Barchiesi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - L Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - M Mazzotta
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - A Venuti
- HPV-UNIT, UOSD Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostic and Technological Innovation (RIDAIT), Translational Research Functional Departmental Area, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - M Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - G Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - G Massimiani
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - D Sergi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - S Carpano
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - P Marchetti
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy.,Medical Oncology Unit B, Policlinico Umberto I, Rome, Italy
| | - S Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I, 'Sapienza' University of Rome, Rome, Italy
| | - T Gamucci
- Medical Oncology, Sandro Pertini Hospital, Rome, Italy
| | - R De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart, Rome, Italy.,Department of Medical Oncology, Policlinico Universitario "A. Gemelli", Rome, Italy
| | - F Tomao
- Department of Gynecology-Obstetrics and Urology, "Sapienza" University of Rome, Rome, Italy
| | - C Natoli
- Department of Medical, Oral and Biotechnological Sciences and Center of Aging Science & Translational Medicine (CeSI-MeT), G. d'Annunzio University, Chieti, Italy
| | - N Tinari
- Department of Medical, Oral and Biotechnological Sciences and Center of Aging Science & Translational Medicine (CeSI-MeT), G. d'Annunzio University, Chieti, Italy
| | - G Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - M Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy.
| | - P Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| |
Collapse
|
164
|
Akin Telli T, Bregni G, Camera S, Deleporte A, Hendlisz A, Sclafani F. PD-1 and PD-L1 inhibitors in oesophago-gastric cancers. Cancer Lett 2019; 469:142-150. [PMID: 31669518 DOI: 10.1016/j.canlet.2019.10.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022]
Abstract
Oesophago-gastric cancers (OGCs) are aggressive tumours. While better peri-operative strategies, increased number of cytotoxic agents and availability of targeted therapies have improved survival, there remains an unmet need for novel treatment approaches. Immune checkpoint inhibitors (ICIs) have marked a new era in cancer management with unprecedented results in several malignancies. Although OGC lagged behind other solid tumours, evidence has increasingly accumulated supporting the contention that modulation of the anti-cancer host immune response may be beneficial for at least some patients. Many trials have been completed in Eastern and Western countries, some of these leading to the approval of ICIs in the refractory setting, and favorable opinion from regulatory agencies is expected also in treatment-naïve, advanced OGC. Furthermore, studies are evaluating ICIs in the early stage setting and exploring the potential of combination treatments. In this article we discuss the biological bases underlying the successful development of ICIs in OGC and review the available data on PD-1 and PD-L1 monoclonal antibodies in this disease. Also, we present ongoing clinical trials of these ICIs that could shape the future treatment landscape of OGC.
Collapse
Affiliation(s)
- Tugba Akin Telli
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giacomo Bregni
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Silvia Camera
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Amelie Deleporte
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alain Hendlisz
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Francesco Sclafani
- Gastrointestinal Unit, Department of Medical Oncology, Institut Jules Bordet - Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
165
|
Page DB, Bear H, Prabhakaran S, Gatti-Mays ME, Thomas A, Cobain E, McArthur H, Balko JM, Gameiro SR, Nanda R, Gulley JL, Kalinsky K, White J, Litton J, Chmura SJ, Polley MY, Vincent B, Cescon DW, Disis ML, Sparano JA, Mittendorf EA, Adams S. Two may be better than one: PD-1/PD-L1 blockade combination approaches in metastatic breast cancer. NPJ Breast Cancer 2019; 5:34. [PMID: 31602395 PMCID: PMC6783471 DOI: 10.1038/s41523-019-0130-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/05/2019] [Indexed: 01/07/2023] Open
Abstract
Antibodies blocking programmed death 1 (anti-PD-1) or its ligand (anti-PD-L1) are associated with modest response rates as monotherapy in metastatic breast cancer, but are generally well tolerated and capable of generating dramatic and durable benefit in a minority of patients. Anti-PD-1/L1 antibodies are also safe when administered in combination with a variety of systemic therapies (chemotherapy, targeted therapies), as well as with radiotherapy. We summarize preclinical, translational, and preliminary clinical data in support of combination approaches with anti-PD-1/L1 in metastatic breast cancer, focusing on potential mechanisms of synergy, and considerations for clinical practice and future investigation.
Collapse
Affiliation(s)
- David B. Page
- Providence Cancer Institute; Earle A. Chiles Research Institute, Portland, OR USA
| | - Harry Bear
- Division of Surgical Oncology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA USA
| | - Sangeetha Prabhakaran
- Department of Surgery, Division of Surgery, University of New Mexico; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM USA
| | | | - Alexandra Thomas
- Wake Forest University School of Medicine, Winston-Salem, NC USA
| | | | | | - Justin M. Balko
- Department of Medicine and Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN USA
| | - Sofia R. Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, MD USA
| | - Rita Nanda
- The University of Chicago, Chicago, IL USA
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | | | - Julia White
- Ohio State Wexner Medical Center, Columbus, OH USA
| | | | | | | | | | - David W. Cescon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON Canada
| | | | - Joseph A. Sparano
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY USA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital; Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, MA USA
| | - Sylvia Adams
- Perlmutter Cancer Center, NYU School of Medicine, New York, NY USA
| |
Collapse
|
166
|
Tuo Z, Zong Y, Li J, Xiao G, Zhang F, Li G, Wang S, Lv Y, Xia J, Liu J. PD-L1 regulation by SDH5 via β-catenin/ZEB1 signaling. Oncoimmunology 2019; 8:1655361. [PMID: 31741753 PMCID: PMC6844322 DOI: 10.1080/2162402x.2019.1655361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 08/02/2019] [Accepted: 08/09/2019] [Indexed: 01/29/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a crucial target for lung cancer immunotherapy. In lung cancer patients with high PD-L1 expression, blocking or reducing its expression can inhibit tumor growth. PD-L1 is regulated by signaling pathways, transcription factors and epigenetic factors, such as the GSK3β/β-catenin pathway, P53 protein and EMT. In our previous study, succinate dehydrogenase 5 (SDH5) was reported to regulate ZEB1 expression, induce EMT and lead to lung cancer metastasis via the GSK3β/β-catenin pathway. It is possible that SDH5 is involved in the mechanisms of PD-L1 regulation.In the present study, we observed a negative correlation between the expression of PD-L1 and SDH5 in vivo and in vitro. The examination of patient tissues also confirmed our results. Furthermore, we also found that SDH5 could reverse PD-L1 expression by the GSK3β/β-catenin/ZEB1 pathways. All these results reveal that SDH5 regulates PD-L1 expression and suggest that SDH5 can be used as a marker to predict tumor immune micro-states and provide guidance for clinical immunotherapy.
Collapse
Affiliation(s)
- Zhan Tuo
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangqin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Furong Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guiling Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sihua Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Lv
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
167
|
Viale G, Morganti S, Ferraro E, Zagami P, Marra A, Curigliano G. What therapies are on the horizon for HER2 positive breast cancer? Expert Rev Anticancer Ther 2019; 19:811-822. [PMID: 31448640 DOI: 10.1080/14737140.2019.1660164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: Despite dramatic improvements in survival achieved with currently available anti-HER2 agents, HER2-positive metastatic breast cancer remains an almost invariably deadly disease, with primary or acquired resistance to HER2-directed agents developing during treatment. Many efforts are focused on identifying new agents that may more effectively inhibit HER2 signaling and on possible combination strategies. Areas covered: This review summarizes the landscape of drugs under development for HER2-positive metastatic breast cancer, as antibody-drug conjugates, monoclonal anti-HER2 antibodies, bispecific antibodies, or novel tyrosine kinase inhibitors. Moreover, available data for possible combination of anti-HER2 drugs and different agents, as immunotherapy, PI3K/mTOR inhibitors, CDK4/6 inhibitors currently under evaluation are reviewed. These strategies may overcome mechanisms of resistance and further improve patient outcomes. Expert opinion: Identification of valuable predictive biomarkers is needed to better inform choice of treatment sequence for the individual patient and limit the financial toxicity of these agents.
Collapse
Affiliation(s)
- Giulia Viale
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Stefania Morganti
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Emanuela Ferraro
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Paola Zagami
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Antonio Marra
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology IRCCS , Milan , Italy.,Department of Oncology and Haematology, University of Milan , Milan , Italy
| |
Collapse
|
168
|
Mittal D, Vijayan D, Neijssen J, Kreijtz J, Habraken MMJM, Van Eenennaam H, Van Elsas A, Smyth MJ. Blockade of ErbB2 and PD-L1 using a bispecific antibody to improve targeted anti-ErbB2 therapy. Oncoimmunology 2019; 8:e1648171. [PMID: 31646095 DOI: 10.1080/2162402x.2019.1648171] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/14/2019] [Accepted: 07/23/2019] [Indexed: 01/04/2023] Open
Abstract
A significant proportion of human epidermal growth factor receptor 2 (Her2/ErbB2)-positive metastatic breast cancer patients are refractory to Her2-targeted trastuzumab-like therapy. Some of this resistance has been attributed to the upregulation of immune checkpoints such as programmed cell death-1 (PD-1) and its ligand, PD-L1 in Her2-positive breast cancer patients. Therefore, therapies targeting both the PD-1/PD-L1 interaction and oncogenic Her2 signaling are of significant clinical interest. Here, we constructed a mouse bispecific antibody targeting PD-L1 and rat Her2 (referred to as BsPD-L1xrErbB2) aiming to redirect the anti-PD-L1 response toward Her2-expressing tumor cells. BsPD-L1xrErbB2 demonstrated additive binding to interferon (IFN)-γ treated Her2+ TUBO tumor cells, but it did not affect the proliferation of tumor cells in-vitro. BsPD-L1xrErbB2 also blocked the PD-1/PD-L1 interaction. This bispecific antibody was constructed with a mouse IgG2a Fc backbone and interacted with Fcγ receptors and resulted in complement deposition (C3). ADCC and complement action could be potential mechanisms of action of this molecule. BsPD-L1xrErbB2 successfully reduced TUBO tumor growth and increased tumor rejection rate compared to the monovalent anti-PD-L1, monovalent anti-ErbB2 or the combination of anti-PD-L1 and anti-ErbB2 monotherapies. The enhanced anti-tumor effect of BsPD-L1xrErbB2 was dependent on CD8+ T lymphocytes and IFN-γ, as depletion of CD8+ T lymphocytes and neutralization of IFN-γ completely abolished the antitumor activity of the bispecific antibody. Consistently, BsPD-L1xrErbB2 treatment also increased the frequency of intratumor CD8+ T lymphocytes. Taken together, our data support a bispecific antibody approach to enhance the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade in Her2-positive metastatic breast cancers.
Collapse
Affiliation(s)
- Deepak Mittal
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
| | - Dipti Vijayan
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
| | | | | | | | | | | | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland Australia
| |
Collapse
|
169
|
Ivashkiv LB. IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat Rev Immunol 2019; 18:545-558. [PMID: 29921905 DOI: 10.1038/s41577-018-0029-z] [Citation(s) in RCA: 838] [Impact Index Per Article: 139.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IFNγ is a cytokine with important roles in tissue homeostasis, immune and inflammatory responses and tumour immunosurveillance. Signalling by the IFNγ receptor activates the Janus kinase (JAK)-signal transducer and activator of transcription 1 (STAT1) pathway to induce the expression of classical interferon-stimulated genes that have key immune effector functions. This Review focuses on recent advances in our understanding of the transcriptional, chromatin-based and metabolic mechanisms that underlie IFNγ-mediated polarization of macrophages to an 'M1-like' state, which is characterized by increased pro-inflammatory activity and macrophage resistance to tolerogenic and anti-inflammatory factors. In addition, I describe the newly discovered effects of IFNγ on other leukocytes, vascular cells, adipose tissue cells, neurons and tumour cells that have important implications for autoimmunity, metabolic diseases, atherosclerosis, neurological diseases and immune checkpoint blockade cancer therapy.
Collapse
Affiliation(s)
- Lionel B Ivashkiv
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA. .,Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
170
|
Chia S, Bedard PL, Hilton J, Amir E, Gelmon K, Goodwin R, Villa D, Cabanero M, Tu D, Tsao M, Seymour L. A Phase Ib Trial of Durvalumab in Combination with Trastuzumab in HER2-Positive Metastatic Breast Cancer (CCTG IND.229). Oncologist 2019; 24:1439-1445. [PMID: 31420468 DOI: 10.1634/theoncologist.2019-0321] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/12/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors are active in a broad range of cancers, including programmed death ligand 1 (PD-L1)-positive, triple-negative, metastatic breast cancer (MBC). Antibody-dependent cell-mediated cytotoxicity is a mechanism of action of trastuzumab. We performed a phase Ib trial of durvalumab and trastuzumab in HER2-positive MBC previously treated with chemotherapy and anti-HER2 antibodies to assess safety, efficacy, and correlative endpoints. PATIENTS AND METHODS Patients with HER2-positive MBC were enrolled on a standard 3 + 3 design. Dose level 1 was durvalumab (1,125 mg intravenously day 1) and trastuzumab (8 mg/kg intravenously loading, then 6 mg/kg day 1) on a q3 weekly cycle. An expansion cohort at the recommended phase II dose (RP2D) performed tumor biopsies at baseline and after cycle 1. The primary endpoint was to establish the RP2D. RESULTS Fifteen patients were accrued from April to December 2016, of which 14 were evaluable for response. Median age was 54 years (range 40-86); the majority had visceral disease (87%) and at least three prior (adjuvant and/or metastatic) lines of chemotherapy (73%), including trastuzumab (93%), pertuzumab (60%), and trastuzumab-emtansine (93%) for MBC. No dose-limiting toxicities were observed at dose level 1 (n = 6) or dose expansion (n = 9) during cycle 1. One patient developed a grade ≥3 immune-related adverse event (grade 4 diabetes mellitus). No responses by RECIST were seen, with 4 of 14 patients (29%) demonstrating stable disease as best response at week 6 (median duration, 2.7 months). All patients had <1% PD-L1 expression on either archival tissue (7/15) or prestudy biopsy (8/15). In the dose expansion cohort, evaluable pretreatment and on-treatment tumor biopsies (n = 5) showed minimal CD8 cell infiltration. CONCLUSION The RP2D of durvalumab and trastuzumab is standard full doses of both agents. No significant clinical activity was observed in patients with heavily pretreated HER2-positive PD-L1-negative MBC. IMPLICATIONS FOR PRACTICE This phase Ib trial with associated correlative endpoints provides insights into the lack of activity of the combination of durvalumab and trastuzumab in heavily pretreated HER2-positive metastatic breast cancer (MBC). No significant clinical activity was observed in patients with heavily pretreated HER2-positive programmed death ligand 1 (PD-L1)-negative MBC with evidence of cytotoxic T-cell exhaustion. Furthermore, all patients had no expression of PD-L1 in the tumor cells. These data support the importance of PD-L1 as an important selection biomarker and the need to assess the tumor microenvironment for immune regulatory cells. Further work is needed to understand how to activate the "cold" tumors to be able to combine current immune-oncology agents.
Collapse
Affiliation(s)
- Stephen Chia
- British Columbia Cancer Agency-Vancouver, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Phillipe L Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - John Hilton
- The Ottawa Hospital Research Institute, Ontario, Canada
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Karen Gelmon
- British Columbia Cancer Agency-Vancouver, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Diego Villa
- British Columbia Cancer Agency-Vancouver, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Cabanero
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group (CCTG), Kingston, Ontario, Canada
| | - Ming Tsao
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lesley Seymour
- Canadian Cancer Trials Group (CCTG), Kingston, Ontario, Canada
| |
Collapse
|
171
|
Kodumudi KN, Ramamoorthi G, Snyder C, Basu A, Jia Y, Awshah S, Beyer AP, Wiener D, Lam L, Zhang H, Greene MI, Costa RLB, Czerniecki BJ. Sequential Anti-PD1 Therapy Following Dendritic Cell Vaccination Improves Survival in a HER2 Mammary Carcinoma Model and Identifies a Critical Role for CD4 T Cells in Mediating the Response. Front Immunol 2019; 10:1939. [PMID: 31475002 PMCID: PMC6702967 DOI: 10.3389/fimmu.2019.01939] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/31/2019] [Indexed: 12/27/2022] Open
Abstract
Patients with metastatic HER2 breast cancer (MBC) often become resistant to HER 2 targeted therapy and have recurrence of disease. The Panacea trial suggested that HER2 MBC patients were more likely to respond to checkpoint therapy if TIL were present or if tumor expressed PD-L1. We assessed whether type I polarized dendritic cells (DC1) could improve checkpoint therapy in a preclinical model of HER2+ breast cancer. TUBO bearing mice were vaccinated with either MHC class I or class II HER2 peptide pulsed DC1 (class I or class II HER2-DC1) concurrently or sequentially with administration of anti-PD-1 or anti-PDL1. Infiltration of tumors by immune cells, induction of anti-HER2 immunity and response to therapy was evaluated. Class I or class II HER2-DC1 vaccinated mice generated anti-HER2 CD8 or CD4+ T cell immune responses and demonstrated delayed tumor growth. Combining both MHC class I and II HER2-pulsed DC1 did not further result in inhibition of tumor growth or enhanced survival compared to individual administration. Interestingly class II HER2-DC1 led to both increased CD4 and CD8 T cells in the tumor microenvironment while class I peptides typically resulted in only increased CD8 T cells. Anti-PD-1 but not anti-PD-L1 administered sequentially with class I or class II HER2-DC1 vaccine could improve the efficacy of HER2-DC1 vaccine as measured by tumor growth, survival, infiltration of tumors by T cells and increase in systemic anti-HER2 immune responses. Depletion of CD4+ T cells abrogated the anti-tumor efficacy of combination therapy with class II HER2-DC1 and anti-PD-1, suggesting that tumor regression was CD4 dependent. Since class II HER2-DC1 was as effective as class I, we combined class II HER2-DC1 vaccine with anti-rat neu antibodies and anti-PD-1 therapy. Combination therapy demonstrated further delay in tumor growth, and enhanced survival compared to control mice. In summary, Class II HER2-DC1 drives both a CD4 and CD8 T cell tumor infiltration that leads to increased survival, and in combination with anti-HER2 therapy and checkpoint blockade can improve survival in preclinical models of HER2 positive breast cancer and warrants exploration in patients with HER2 MBC.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/therapy
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Combined Modality Therapy
- Dendritic Cells/immunology
- Female
- Humans
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Transgenic
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Rats
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Survival Analysis
- Treatment Outcome
- Tumor Burden/drug effects
- Tumor Burden/immunology
Collapse
Affiliation(s)
- Krithika N. Kodumudi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Ganesan Ramamoorthi
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Colin Snyder
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Amrita Basu
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Yongsheng Jia
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Sabrina Awshah
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Amber P. Beyer
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Doris Wiener
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Lian Lam
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongtao Zhang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark I. Greene
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ricardo L. B. Costa
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| | - Brian J. Czerniecki
- Clinical Science & Immunology Program, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
172
|
Liu Z, Han C, Dong C, Shen A, Hsu E, Ren Z, Lu C, Liu L, Zhang A, Timmerman C, Pu Y, Wang Y, Chen M, Qiao J, Fu YX. Hypofractionated EGFR tyrosine kinase inhibitor limits tumor relapse through triggering innate and adaptive immunity. Sci Immunol 2019; 4:eaav6473. [PMID: 31399492 DOI: 10.1126/sciimmunol.aav6473] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 03/05/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022]
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are a first-line therapy for rapidly killing tumors such as those associated with non-small cell lung cancer by blocking oncogenic receptor signaling, but tumor relapse often occurs. Here, we have observed that hypofractionated EGFR TKI treatment (HypoTKI) is more potent than standard hyperfractionated EGFR TKI treatment (HyperTKI), and its antitumor effect associated with preventing tumor relapse depends on T cells. HypoTKI triggers greater innate sensing for type I IFN and CXCL10 production through the Myd88 signaling pathway to enhance tumor-specific T cell infiltration and reactivation. We also demonstrate that timely programmed cell death ligand-1 (PD-L1) blockade can synergize with HypoTKI to control advanced large tumors and effectively limit tumor relapse without severe side effects. Our study provides evidence for exploring the potential of a proper combination of EGFR TKIs and immunotherapy as a first-line treatment for treating EGFR-driven tumors.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Animals
- Antibodies/drug effects
- Antibodies/immunology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/metabolism
- Female
- Immunity, Innate/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Zhida Liu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Chuanhui Han
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Chunbo Dong
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Aijun Shen
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Eric Hsu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Zhenhua Ren
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Changzheng Lu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Longchao Liu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Anli Zhang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Casey Timmerman
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang Pu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Mingyi Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Jian Qiao
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang-Xin Fu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75235, USA.
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
173
|
Criscitiello C, Curigliano G. Treatment in real-life patients with HER2-positive metastatic breast cancer: What we learn from the KAMILLA trial? Eur J Cancer 2019; 117:1-4. [DOI: 10.1016/j.ejca.2019.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
|
174
|
Wu S, Zhang Q, Zhang F, Meng F, Liu S, Zhou R, Wu Q, Li X, Shen L, Huang J, Qin J, Ouyang S, Xia Z, Song H, Feng XH, Zou J, Xu P. HER2 recruits AKT1 to disrupt STING signalling and suppress antiviral defence and antitumour immunity. Nat Cell Biol 2019; 21:1027-1040. [DOI: 10.1038/s41556-019-0352-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 06/03/2019] [Indexed: 12/19/2022]
|
175
|
Planes-Laine G, Rochigneux P, Bertucci F, Chrétien AS, Viens P, Sabatier R, Gonçalves A. PD-1/PD-L1 Targeting in Breast Cancer: The First Clinical Evidences Are Emerging. A Literature Review. Cancers (Basel) 2019; 11:E1033. [PMID: 31336685 PMCID: PMC6679223 DOI: 10.3390/cancers11071033] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Recently, the development of immunotherapy through the immune checkpoint blockade led to long-lasting responses in several types of cancers that are refractory to conventional treatments, such as melanoma or non-small cell lung cancer. Immunotherapy has also demonstrated significant improvements in various other types of cancers. However, breast cancer remains one of the tumors that have not experienced the explosion of immunotherapy yet. Indeed, breast cancer was traditionally considered as being weakly immunogenic with a lower mutational load compared to other tumor types. In the last few years, anti-PD1/PD-L1 (Programmed death-ligand 1) agents have been evaluated in breast cancer, particularly in the triple negative subtype, with promising results observed when delivered as monotherapy or in combination with conventional treatments. In this review, we will report the results of the most recent studies evaluating immune checkpoint inhibitors in breast cancer. In addition, we will discuss the concomitant development of possible biomarkers, which is required for improving the selection of patients with the highest probability of benefiting from these agents.
Collapse
Affiliation(s)
- Gabrielle Planes-Laine
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Philippe Rochigneux
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
- CRCM-Tumor Immunology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - François Bertucci
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
- CRCM-Predictive Oncology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Anne-Sophie Chrétien
- CRCM-Tumor Immunology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Patrice Viens
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Renaud Sabatier
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
- CRCM-Predictive Oncology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France
| | - Anthony Gonçalves
- Department of Medical Oncology, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France.
- CRCM-Predictive Oncology laboratory, Aix-Marseille University, Inserm U1068, CNRS UMR7258, Institute Paoli-Calmettes, 13009 Marseille, France.
| |
Collapse
|
176
|
Resistance mechanisms to anti-HER2 therapies in HER2-positive breast cancer: Current knowledge, new research directions and therapeutic perspectives. Crit Rev Oncol Hematol 2019; 139:53-66. [DOI: 10.1016/j.critrevonc.2019.05.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/19/2018] [Accepted: 05/01/2019] [Indexed: 01/10/2023] Open
|
177
|
Nicolini A, Barak V, Biava P, Ferrari P, Rossi G, Carpi A. The Use of Immunotherapy to Treat Metastatic Breast Cancer. Curr Med Chem 2019; 26:941-962. [PMID: 29424297 DOI: 10.2174/0929867325666180209124052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/21/2022]
Abstract
This article reviews the principal attempts of immune-modulation or immune therapy in metastatic breast cancer. It considers their rationale and reports on results from the relevant key clinical trials. Immune-modulatory or immune-stimulating cytokines used alone or combined with conventional therapies is among the principal approaches of immune manipulation in breast cancer. As this issue has recently been reviewed by us, the aim of the current article is to discuss our updated and unpublished data on this topic. Overall survival in luminal (28 patients) and non-luminal (9 patients) molecular subtypes is 91 and 59 months respectively that is about two and half or three times longer than expected. Thereafter, we focus on monoclonal antibodies (mAb) based-therapies including novel strategies to overcome resistance to anti-HER2 mAb. The main vaccine platforms in different molecular subtypes and immune therapies in triple negative metastatic breast cancer (m-TNBC) are discussed in the last sections. Some phase III investigations have already changed the current clinical practice. In fact, pertuzumab plus trastuzumab and docetaxel is the recommended first line regimen in HER2 positive locally recurrent or metastatic breast cancer and bevacizumab plus paclitaxel or docetaxel is a reasonable option for m-TNBC. In some other observational or phase I/II studies on first-line trastuzumab plus chemotherapy and hormonal therapy and in that on HER2 peptide/protein vaccines promising although preliminary findings have been reported to be further validated. In the remaining studies, results were disappointing. In the future, finding new predictive biomarkers and exploring more suitable synergizing combinations, time and dose-dependent-scheduled sequences of currently and further investigated immunological approaches are main challenges.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Italy
| | - Vivian Barak
- Immunology Lab for tumor diagnosis, Hadassah University, Jerusalem, Israel
| | - Piermario Biava
- Scientific Institute of Research and Care Multimedica, Milan, Italy
| | - Paola Ferrari
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Italy
| | - Giuseppe Rossi
- Unit of Epidemiology and Biostatistics, Institute of Clinical Physiology, National Council of Research, Pisa, Italy
| | - Angelo Carpi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
178
|
Gombos A, Franzoi MA, Awada A. Investigational drugs in early stage clinical trials for the treatment of HER2+ breast cancer. Expert Opin Investig Drugs 2019; 28:617-627. [DOI: 10.1080/13543784.2019.1633306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Andrea Gombos
- Department of Oncology Medicine, Institut Jules Bordet, Brussels, Belgium
| | - Maria Alice Franzoi
- Clinical Trial Support Unit, Institut Jules Bordet Institute and L’Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Ahmad Awada
- Department of Oncology Medicine, Institut Jules Bordet, Brussels, Belgium
- Department of Oncology Medicine, Université Libre de Bruxelles, Bruxelles, Belgium
| |
Collapse
|
179
|
Muntasell A, Servitja S, Cabo M, Bermejo B, Pérez-Buira S, Rojo F, Costa-García M, Arpí O, Moraru M, Serrano L, Tusquets I, Martínez MT, Heredia G, Vera A, Martínez-García M, Soria L, Comerma L, Santana-Hernández S, Eroles P, Rovira A, Vilches C, Lluch A, Albanell J, López-Botet M. High Numbers of Circulating CD57 + NK Cells Associate with Resistance to HER2-Specific Therapeutic Antibodies in HER2 + Primary Breast Cancer. Cancer Immunol Res 2019; 7:1280-1292. [PMID: 31189644 DOI: 10.1158/2326-6066.cir-18-0896] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/11/2019] [Accepted: 06/10/2019] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cells can orchestrate effective antitumor immunity. The presence of tumor-infiltrating NK cells in diagnostic biopsies predicts pathologic complete response (pCR) to HER2-specific therapeutic antibodies in patients with primary breast cancer. Here, we analyzed whether diversity in circulating NK cells might influence tumor infiltration and HER2-specific therapeutic antibody efficacy. We found that numbers of circulating CD57+ NK cells inversely correlated with pCR to HER2-specific antibody treatment in patients with primary breast cancer independently of age, traditional clinicopathologic factors, and CD16A 158F/V genotype. This association was uncoupled from the expression of other NK-cell receptors, the presence of adaptive NK cells, or changes in major T-cell subsets, reminiscent of cytomegalovirus-induced immunomodulation. NK-cell activation against trastuzumab-coated HER2+ breast cancer cells was comparable in patients with high and low proportions of CD57+ NK cells. However, circulating CD57+ NK cells displayed decreased CXCR3 expression and CD16A-induced IL2-dependent proliferation in vitro Presence of CD57+ NK cells was reduced in breast tumor-associated infiltrates as compared with paired peripheral blood samples, suggesting deficient homing, proliferation, and/or survival of NK cells in the tumor niche. Indeed, numbers of circulating CD57+ were inversely related to tumor-infiltrating NK-cell numbers. Our data reveal that NK-cell differentiation influences their antitumor potential and that CD57+ NK cells may be a biomarker useful for tailoring HER2 antibody-based therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Aura Muntasell
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.
| | - Sònia Servitja
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Mariona Cabo
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Begoña Bermejo
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain
| | - Sandra Pérez-Buira
- Department of Pathology, IIS "Fundacion Jimenez Diaz University Hospital," Madrid, Spain
| | - Federico Rojo
- Department of Pathology, IIS "Fundacion Jimenez Diaz University Hospital," Madrid, Spain
| | | | - Oriol Arpí
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Manuela Moraru
- HLA-Immunogenetics Department, Instituto Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Laia Serrano
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Ignasi Tusquets
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | - Andrea Vera
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - María Martínez-García
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Laura Soria
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Laura Comerma
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Sara Santana-Hernández
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Pilar Eroles
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain.,Biomedical Research Institute, INCLIVA, Valencia, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Carlos Vilches
- HLA-Immunogenetics Department, Instituto Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Ana Lluch
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain.,Biomedical Research Institute, INCLIVA, Valencia, Spain.,Universitat de Valencia, Valencia, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain. .,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Miguel López-Botet
- Immunity and Infection, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
180
|
Bayraktar S, Batoo S, Okuno S, Glück S. Immunotherapy in breast cancer. J Carcinog 2019; 18:2. [PMID: 31160888 PMCID: PMC6540776 DOI: 10.4103/jcar.jcar_2_19] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/09/2019] [Indexed: 12/31/2022] Open
Abstract
The idea of using the immune system to fight cancer is over 100 years old. A new molecular approach led to a better understanding of the immune system. Checkpoint regulation, understanding the roles of Tregs, Th1, and Th2, development of Chimeric antigen receptor (CAR)-T cells, as well as regulation of dendritic cells and macrophages, are just a few examples of our understating that has also led to the discovery of immune checkpoint inhibitors (ICIs) and modulators. This led the Nobel Prize committee in 2018, to award Dr. James P. Allison the Nobel Prize in medicine for the discovery of Cytotoxic T-lymphocyte-associated antigen-4, and Dr. Tasuku Honjo for the discovery of programmed cell death-1 (PD-1)/PD-1-ligand (PDL-1). Several ICIs are already approved by the regulatory authorities, and many more are currently used in studies of several solid tumors and hematologic malignancies. Positive studies have led to the US Food and Drug Administration (FDA) and European Medicines Agency approval of a number of these compounds, but none to date are approved in breast cancer (BC). Moreover, PD-1/PDL-1, MSI high (and dMMR), and tumor mutational burden are the currently “best” predictive markers for benefit from immunotherapy. BCs have some of these markers positive only in subsets but less frequently expressed than most other solid tumors, for example, malignant melanoma or non-small cell lung cancer. To improve the potential efficacy of ICI in BC, the addition of chemotherapy was one of the strategies. Many early and large clinical trials in all phases are underway in BC. We will discuss the role of immune system in BC editing, and the potential impact of immunotherapy in BC outcomes.
Collapse
Affiliation(s)
- Soley Bayraktar
- Department of Medicine, Division of Medical Oncology and Hematology, Mayo Clinic Health System, Eau Claire, WI, USA.,Department of Medicine, Division of Medical Oncology and Hematology, Biruni University School of Medicine, Istanbul, Turkey
| | - Sameer Batoo
- Department of Medicine, Division of Medical Oncology and Hematology, Mayo Clinic Health System, Eau Claire, WI, USA
| | - Scott Okuno
- Department of Medicine, Division of Medical Oncology and Hematology, Mayo Clinic Health System, Eau Claire, WI, USA
| | - Stefan Glück
- Vice President Global Medical Affairs, Early Assets, Celgene Corporation, Summit, NJ, USA
| |
Collapse
|
181
|
Chu DT, Bac ND, Nguyen KH, Tien NLB, Thanh VV, Nga VT, Ngoc VTN, Anh Dao DT, Hoan LN, Hung NP, Trung Thu NT, Pham VH, Vu LN, Pham TAV, Thimiri Govinda Raj DB. An Update on Anti-CD137 Antibodies in Immunotherapies for Cancer. Int J Mol Sci 2019; 20:ijms20081822. [PMID: 31013788 PMCID: PMC6515339 DOI: 10.3390/ijms20081822] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/05/2019] [Accepted: 04/10/2019] [Indexed: 12/16/2022] Open
Abstract
The selective expression of CD137 on cells of the immune system (e.g., T and DC cells) and oncogenic cells in several types of cancer leads this molecule to be an attractive target to discover cancer immunotherapy. Therefore, specific antibodies against CD137 are being studied and developed aiming to activate and enhance anti-cancer immune responses as well as suppress oncogenic cells. Accumulating evidence suggests that anti-CD137 antibodies can be used separately to prevent tumor in some cases, while in other cases, these antibodies need to be co-administered with other antibodies or drugs/vaccines/regents for a better performance. Thus, in this work, we aim to update and discuss current knowledge about anti-cancer effects of anti-CD137 antibodies as mono- and combined-immunotherapies.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam.
- Institute of Cancer Research, Oslo University Hospital, 0372 Oslo, Norway.
| | - Nguyen Duy Bac
- Department of Education and Training, Vietnam Military Medical University, Hanoi 100000, Vietnam.
| | - Khanh-Hoang Nguyen
- National Food Institute, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam.
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam.
| | - Vu Thi Nga
- Institute for Research and Development, Duy Tan University, 03 Quang Trung, Danang 550000, Vietnam.
| | - Vo Truong Nhu Ngoc
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam.
| | - Duong Thi Anh Dao
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
| | - Le Ngoc Hoan
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
| | - Nguyen Phuc Hung
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
| | - Nguyen Thi Trung Thu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam.
| | - Van-Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Le Nguyen Vu
- Organ Transplantation Center, Viet Duc Hospital, Hanoi 100000, Vietnam.
| | - Thuy Anh Vu Pham
- Faculty of Odonto-Stomatology, University of Medicine and Pharmacy, Ho Chi Minh City 700000, Vietnam.
| | | |
Collapse
|
182
|
Eiger D, Pondé NF, de Azambuja E. Pertuzumab in HER2-positive early breast cancer: current use and perspectives. Future Oncol 2019; 15:1823-1843. [PMID: 30938542 DOI: 10.2217/fon-2018-0896] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Although the prognosis of HER2-positive breast cancer patients has dramatically improved with modern chemotherapy and the monoclonal antibody trastuzumab, up to 31% of them will experience a recurrence in the long term. After the unprecedented benefit in overall survival with the addition of the second monoclonal antibody pertuzumab for patients with metastatic disease, the drug was tested with various degrees of success in the preoperative and postoperative settings. In this review, we will focus on the pharmacologic aspects of the drug, including mechanism of action and toxicities, and discuss clinical data regarding its use in advanced and early stage HER2-positive breast cancer, placing in perspective the pros and cons regarding other available drugs and biomarkers.
Collapse
Affiliation(s)
- Daniel Eiger
- Institut Jules Bordet - Université Libre de Bruxelles (ULB); Medical Oncology Department, Academic Promoting Team at Boulevard de Waterloo, 121, 1000, Brussels, Belgium
| | - Noam Falbel Pondé
- Institut Jules Bordet - Université Libre de Bruxelles (ULB); Medical Oncology Department, Academic Promoting Team at Boulevard de Waterloo, 121, 1000, Brussels, Belgium
| | - Evandro de Azambuja
- Institut Jules Bordet - Université Libre de Bruxelles (ULB); Medical Oncology Department, Academic Promoting Team at Boulevard de Waterloo, 121, 1000, Brussels, Belgium
| |
Collapse
|
183
|
Barroso-Sousa R, Barry WT, Guo H, Dillon D, Tan YB, Fuhrman K, Osmani W, Getz A, Baltay M, Dang C, Yardley D, Moy B, Marcom PK, Mittendorf EA, Krop IE, Winer EP, Tolaney SM. The immune profile of small HER2-positive breast cancers: a secondary analysis from the APT trial. Ann Oncol 2019; 30:575-581. [PMID: 30753274 PMCID: PMC8033534 DOI: 10.1093/annonc/mdz047] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Previous data suggest that the immune microenvironment plays a critical role in human epidermal growth factor receptor 2 (HER2) -positive breast cancer; however, there is little known about the immune profiles of small HER2-positive tumors. In this study, we aimed to characterize the immune microenvironment of small HER2-positive breast cancers included in the Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer (APT) trial and to correlate the immune markers with pathological and molecular tumor characteristics. PATIENTS AND METHODS The APT trial was a multicenter, single-arm, phase II study of paclitaxel and trastuzumab in patients with node-negative HER2-positive breast cancer. The study included 406 patients with HER2-positive, node-negative breast cancer, measuring up to 3 cm. Exploratory analysis of tumor infiltrating lymphocytes (TIL), programmed death-ligand 1 (PD-L1) expression (by immunohistochemistry), and immune gene signatures using data generated by nCounter PanCancer Pathways Panel (NanoString Technologies, Seattle, WA), and their association with pathological and molecular characteristics was carried out. RESULTS Of the 406 patients, 328 (81%) had at least one immune assay carried out: 284 cases were evaluated for TIL, 266 for PD-L1, and 213 for immune gene signatures. High TIL (≥60%) were seen with greater frequency in hormone-receptor (HR) negative, histological grades 2 and 3, as well in HER2-enriched and basal-like tumors. Lower stromal PD-L1 (≤1%) expression was seen with greater frequency in HR-positive, histological grade 1, and in luminal tumors. Both TIL and stromal PD-L1 were positively correlated with 10 immune cell signatures, including Th1 and B cell signatures. Luminal B tumors were negatively correlated with those signatures. Significant correlation was seen among these immune markers; however, the magnitude of correlation did not indicate a monotonic relationship between them. CONCLUSION Immune profiles of small HER2-positive breast cancers differ according to HR status, histological grade, and molecular subtype. Further work is needed to explore the implication of these findings on disease outcome. CLINICAL TRIAL REGISTRATION clinicaltrials.gov identifier: NCT00542451.
Collapse
Affiliation(s)
| | - W T Barry
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston
| | - H Guo
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston
| | - D Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - Y B Tan
- Department of Pathology, Brigham and Women's Hospital, Boston
| | | | | | - A Getz
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - M Baltay
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - C Dang
- Breast Cancer Medicine Service, Department of Medicine, Solid Tumor Division, Memorial Sloan Kettering Cancer Center, New York; Department of Medicine, Weill Cornell Medical Center, New York
| | | | - B Moy
- Department of Hematology-Oncology, Massachusetts General Hospital, Boston
| | - P K Marcom
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Durham
| | - E A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston; Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| | | | | | | |
Collapse
|
184
|
Shukla S. A Viral Nanoparticle Cancer Vaccine Delays Tumor Progression and Prolongs Survival in a HER2 + Tumor Mouse Model. ADVANCED THERAPEUTICS 2019; 2:1800139. [PMID: 33855164 PMCID: PMC8043622 DOI: 10.1002/adtp.201800139] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Indexed: 12/17/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression is associated with aggressive tumors with increased incidence of metastasis and recurrence. Therapeutic antibodies such as Trastuzumab inhibit tumor growth through blockade of HER2 receptors. However, the short lifespan of such therapeutic antibodies necessitates repeat administrations with ensuing cardiac toxicity and development of resistance, while offering no protection against relapse. Cancer vaccines targeting HER2 can overcome these shortcomings of passive immunotherapy by instigating an endogenous and sustained immune response and memory against the cancer antigen. The efficacy of a viral nanoparticle (VNP)-based cancer vaccine is demonstrated here in activating a potent anti-HER2 immune response that delays progression of primary tumors as well as metastases and prolongs survival in mice. The results illustrate that the VNP-based vaccine instigates HER2-specific antibodies as well as effector and memory T cells, which contributes to the effectiveness of the vaccine. Given the highly aggressive course of HER2+ cancers, inhibition of disease progression by such cancer vaccines could provide a critical window for interventions with other adjuvant therapies. Moreover, the immune memory generated by this viral nanoparticle-based cancer vaccine could mitigate relapse of the disease.
Collapse
|
185
|
Griguolo G, Pascual T, Dieci MV, Guarneri V, Prat A. Interaction of host immunity with HER2-targeted treatment and tumor heterogeneity in HER2-positive breast cancer. J Immunother Cancer 2019; 7:90. [PMID: 30922362 PMCID: PMC6439986 DOI: 10.1186/s40425-019-0548-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/27/2019] [Indexed: 12/18/2022] Open
Abstract
Growing evidence suggests a clear role of the host immune system in HER2+ breast cancer. In addition, HER2+ breast cancer is generally considered more immunogenic than hormone receptor-positive (HR+)/HER2-, and specific molecular HER2+ subgroups (e.g. HER2-enriched disease) are more immunogenic than others (e.g. Luminal A or B). From a clinical perspective, the immune system plays a relevant prognostic role in HER2+ breast cancer and contributes to the therapeutic effects of trastuzumab. However, as more HER2-targeted agents become available, a better understanding of the role played by the immune system in modulating therapy response to different agents will be needed. Furthermore, the recent introduction in oncology of immune checkpoint inhibitors capable of unleashing anti-tumor immune response opens new possibilities for therapeutic combinations in HER2+ breast cancer. Here, we review the current pre-clinical and clinical data on the interplay between the immune system and HER2+ breast cancer, focusing on different HER2-targeted treatments and the biological heterogeneity that exists within HER2+ disease. Finally, we discuss new therapeutic approaches exploiting the immune system to increase activity or revert resistance to HER2-targeted agents.
Collapse
Affiliation(s)
- Gaia Griguolo
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Tomás Pascual
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain.
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.
| |
Collapse
|
186
|
Pernas S, Tolaney SM. HER2-positive breast cancer: new therapeutic frontiers and overcoming resistance. Ther Adv Med Oncol 2019; 11:1758835919833519. [PMID: 30911337 PMCID: PMC6425535 DOI: 10.1177/1758835919833519] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/23/2019] [Indexed: 12/29/2022] Open
Abstract
The introduction of anti-HER2 therapies to the treatment of patients with HER2-positive breast cancer has led to dramatic improvements in survival in both early and advanced settings. Despite this breakthrough, nearly all patients with metastatic HER2-positive breast cancer eventually progress on anti-HER2 therapy due to de novo or acquired resistance. A better understanding not only of the underlying mechanisms of HER2 therapy resistance but of tumor heterogeneity as well as the host and tumor microenvironment is essential for the development of new strategies to further improve patient outcomes. One strategy has focused on inhibiting the HER2 signaling pathway more effectively with dual-blockade approaches and developing improved anti-HER2 therapies like antibody-drug conjugates, new anti-HER2 antibodies, bispecific antibodies, or novel tyrosine kinase inhibitors that might replace or be used in addition to some of the current anti-HER2 treatments. Combinations of anti-HER2 therapy with other agents like immune checkpoint inhibitors, CDK4/6 inhibitors, and PI3K/AKT/mTOR inhibitors are also being extensively evaluated in clinical trials. These add-on strategies of combining optimized targeted therapies could potentially improve outcomes for patients with HER2-positive breast cancer but may also allow de-escalation of treatment in some patients, potentially sparing some from unnecessary treatments, and their related toxicities and costs.
Collapse
Affiliation(s)
- Sonia Pernas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medical Oncology-Breast Cancer Unit, Institut Catala d’Oncologia (ICO)-H.U. Bellvitge-IDIBELL, Barcelona, Spain
| | - Sara M. Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
187
|
Wu C, Li M, Meng H, Liu Y, Niu W, Zhou Y, Zhao R, Duan Y, Zeng Z, Li X, Li G, Xiong W, Zhou M. Analysis of status and countermeasures of cancer incidence and mortality in China. SCIENCE CHINA-LIFE SCIENCES 2019; 62:640-647. [PMID: 30900169 DOI: 10.1007/s11427-018-9461-5] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022]
Abstract
Cancer is the leading cause of human deaths in the world and produces serious economic burdens. On September 12, 2018, the academic journal A Cancer Journal for Clinicians published an article about the latest statistics of cancers worldwide, which provided a status report on the global burden of 36 cancers in 185 countries worldwide. Cancer has also become a serious public health problem in China and caused more and more attention of the government and people in recent years. This review analyzes the incidence, mortality and prevalent trend of cancers in China, discusses the reasons behind this status, and reviews the potential countermeasures for cancer prevention and control in China.
Collapse
Affiliation(s)
- Chunchun Wu
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Mengna Li
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Hanbing Meng
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Yukun Liu
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Weihong Niu
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Yao Zhou
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Ran Zhao
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Yumei Duan
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Zhaoyang Zeng
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Xiaoling Li
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Guiyuan Li
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China
| | - Ming Zhou
- Key Laboratory of Carcinogenesis of Ministry of Health of China, Xiangya Hospital, Central South University, Changsha, 410078, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education of China, Cancer Research Institute and School of Basic Medicine Science, Central South University, Changsha, 410078, China.
| |
Collapse
|
188
|
Loi S, Giobbie-Hurder A, Gombos A, Bachelot T, Hui R, Curigliano G, Campone M, Biganzoli L, Bonnefoi H, Jerusalem G, Bartsch R, Rabaglio-Poretti M, Kammler R, Maibach R, Smyth MJ, Di Leo A, Colleoni M, Viale G, Regan MM, André F. Pembrolizumab plus trastuzumab in trastuzumab-resistant, advanced, HER2-positive breast cancer (PANACEA): a single-arm, multicentre, phase 1b-2 trial. Lancet Oncol 2019; 20:371-382. [PMID: 30765258 DOI: 10.1016/s1470-2045(18)30812-x] [Citation(s) in RCA: 338] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND HER2-positive breast cancers usually contain large amounts of T-cell infiltrate. We hypothesised that trastuzumab resistance in HER2-positive breast cancer could be mediated by immune mechanisms. We assessed the safety and anti-tumour activity of pembrolizumab, a programmed cell death protein 1 (PD-1) inhibitor, added to trastuzumab in trastuzumab-resistant, advanced HER2-positive breast cancer. METHODS We did this single-arm, multicentre, phase 1b-2 trial in 11 centres based in five countries. Eligible participants were women aged 18 years or older, who had advanced, histologically confirmed, HER2-positive breast cancer; documented progression during previous trastuzumab-based therapy; an Eastern Cooperative Oncology Group performance status of 0 or 1; and a formalin-fixed, paraffin-embedded metastatic tumour biopsy for central assessment of programmed cell death 1 ligand 1 (PD-L1) status. In phase 1b, we enrolled patients with PD-L1-positive tumours in a 3 + 3 dose-escalation of intravenous pembrolizumab (2 mg/kg and 10 mg/kg, every 3 weeks) plus 6 mg/kg of intravenous trastuzumab. The primary endpoint of the phase 1b study was the incidence of dose-limiting toxicity and recommended phase 2 dose; however, a protocol amendment on Aug 28, 2015, stipulated a flat dose of pembrolizumab of 200 mg every 3 weeks in all Merck-sponsored trials. In phase 2, patients with PD-L1-positive and PD-L1-negative tumours were enrolled in parallel cohorts and received the flat dose of pembrolizumab plus standard trastuzumab. The primary endpoint of the phase 2 study was the proportion of PD-L1-positive patients achieving an objective response. This trial is registered in ClinicalTrials.gov, number NCT02129556, and with EudraCT, number 2013-004770-10, and is closed. FINDINGS Between Feb 2, 2015, and April 5, 2017, six patients were enrolled in phase 1b (n=3 received 2 mg/kg pembrolizumab, n=3 received 10 mg/kg pembrolizumab) and 52 patients in phase 2 (n=40 had PD-L1-positive tumours, n=12 had PD-L1-negative tumours). The data cutoff for this analysis was Aug 7, 2017. During phase 1b, there were no dose-limiting toxicities in the dose cohorts tested. Median follow-up for the phase 2 cohort was 13·6 months (IQR 11·6-18·4) for patients with PD-L1-positive tumours, and 12·2 months (7·9-12·2) for patients with PD-L1-negative tumours. Six (15%, 90% CI 7-29) of 40 PD-L1-positive patients achieved an objective response. There were no objective responders among the PD-L1-negative patients. The most common treatment-related adverse event of any grade was fatigue (12 [21%] of 58 patients). Grade 3-5 adverse events occurred in 29 (50%) of patients, treatment-related grade 3-5 adverse events occurred in 17 (29%), and serious adverse events occurred in 29 (50%) patients. The most commonly occurring serious adverse events were dyspnoea (n=3 [5%]), pneumonitis (n=3 [5%]), pericardial effusion (n=2 [3%]), and upper respiratory infection (n=2 [3%]). There was one treatment-related death due to Lambert-Eaton syndrome in a PD-L1-negative patient during phase 2. INTERPRETATION Pembrolizumab plus trastuzumab was safe and showed activity and durable clinical benefit in patients with PD-L1-positive, trastuzumab-resistant, advanced, HER2-positive breast cancer. Further studies in this breast cancer subtype should focus on a PD-L1-positive population and be done in less heavily pretreated patients. FUNDING Merck, International Breast Cancer Study Group.
Collapse
Affiliation(s)
- Sherene Loi
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia.
| | - Anita Giobbie-Hurder
- IBCSG Statistical Center, Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Rina Hui
- Westmead Hospital and the University of Sydney, Sydney, NSW, Australia
| | - Giuseppe Curigliano
- University of Milano, Milan, Italy; IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Mario Campone
- Institut de Cancérologie de l'Ouest, Saint-Herblain, Nantes, France
| | | | - Hervé Bonnefoi
- Institut Bergonié Comprehensive Cancer Center, Université de Bordeaux, Bordeaux, France
| | - Guy Jerusalem
- International Breast Cancer Study Group, CHU Liège, Liège University, Liège, Belgium
| | | | - Manuela Rabaglio-Poretti
- University Hospital Inselspital, Bern, Switzerland; International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Roswitha Kammler
- International Breast Cancer Study Group Coordinating Center, Bern, Switzerland; International Breast Cancer Study Group and Central Pathology Office, Bern, Switzerland
| | - Rudolf Maibach
- International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Angelo Di Leo
- International Breast Cancer Study Group and Ospedale di Prato-AUSL Toscana Centro, Prato, Italy
| | - Marco Colleoni
- International Breast Cancer Study Group and Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Viale
- University of Milano, Milan, Italy; IEO, European Institute of Oncology IRCCS, Milan, Italy; International Breast Cancer Study Group and Central Pathology Office, Bern, Switzerland
| | - Meredith M Regan
- IBCSG Statistical Center, Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Fabrice André
- Institut Gustave Roussy, Université Paris Sud, INSERM U981, Villejuif, France
| |
Collapse
|
189
|
König L, Mairinger FD, Hoffmann O, Bittner AK, Schmid KW, Kimmig R, Kasimir-Bauer S, Bankfalvi A. Dissimilar patterns of tumor-infiltrating immune cells at the invasive tumor front and tumor center are associated with response to neoadjuvant chemotherapy in primary breast cancer. BMC Cancer 2019; 19:120. [PMID: 30717704 PMCID: PMC6360695 DOI: 10.1186/s12885-019-5320-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 01/25/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) are described as an important immune modulator in the tumor microenvironment and are associated with breast cancer (BC) outcome. The spatial analysis of TILs and TIL subtype distribution at the invasive tumor front (ITF) and the tumor center (TC) might provide further insights into tumor progression. METHODS We analyzed core biopsies from 87 pre-therapeutic BC patients for total TILs and the following subtypes: CD3+, CD4+, CD8+, CD20+ and CD68+ cells in correlation to clinicopathological parameters and disseminated tumor cells (DTCs) in the bone marrow. RESULTS TILs and TIL subtypes showed significantly different spatial distribution among both tumor areas. TILs, especially CD3+ T cells were associated with the tumor status and tumor grading. BC patients responding to neoadjuvant chemotherapy had significantly more TILs and CD3+ T cells at the TC. The presence of DTCs after NACT was related to CD4+ infiltration at the TC. CONCLUSION The dissimilar spatial association of TILs and TIL subtypes with clinicopathological parameters, NACT response and minimal residual disease underlines the necessity of detailed TIL analysis for a better understanding of immune modulatory processes.
Collapse
Affiliation(s)
- Lisa König
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Fabian D. Mairinger
- Institute for Pathology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Oliver Hoffmann
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Ann-Kathrin Bittner
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Kurt W. Schmid
- Institute for Pathology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Agnes Bankfalvi
- Institute for Pathology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
190
|
Targeting Multiple Receptors to Increase Checkpoint Blockade Efficacy. Int J Mol Sci 2019; 20:ijms20010158. [PMID: 30621125 PMCID: PMC6337574 DOI: 10.3390/ijms20010158] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/30/2022] Open
Abstract
Immune checkpoint blockade therapy is a powerful treatment strategy for many cancer types. Many patients will have limited responses to monotherapy targeted to a single immune checkpoint. Both inhibitory and stimulatory immune checkpoints continue to be discovered. Additionally, many receptors previously identified to play a role in tumor formation and progression are being found to have immunomodulatory components. The success of immunotherapy depends on maximizing pro-anti-tumor immunity while minimizing immunosuppressive signaling. Combining immune checkpoint targeted approaches with each other or with other receptor targets is a promising schema for future therapeutic regimen designs.
Collapse
|
191
|
|
192
|
Aydiner A. Systemic Treatment of HER2-Overexpressing Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
193
|
Masu T, Atsukawa M, Nakatsuka K, Shimizu M, Miura D, Arai T, Harimoto H, Kondo C, Kaneko K, Futagami S, Kawamoto C, Takahashi H, Iwakiri K. Anti-CD137 monoclonal antibody enhances trastuzumab-induced, natural killer cell-mediated cytotoxicity against pancreatic cancer cell lines with low human epidermal growth factor-like receptor 2 expression. PLoS One 2018; 13:e0200664. [PMID: 30596643 PMCID: PMC6312288 DOI: 10.1371/journal.pone.0200664] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
Abstract
Because human epidermal growth factor-like receptor (HER) 2 is expressed on the surface of human pancreatic carcinoma cells to varying degrees, trastuzumab, an anti-HER2 monoclonal antibody (mAb), is expected to exert antibody-dependent, natural killer (NK) cell-mediated cytotoxicity (ADCC) against the cells. However, some reports found that the effect of trastuzumab against human pancreatic carcinoma cells was limited because most express only limited HER2. We examined whether anti-CD137 stimulating mAb could enhance trastuzumab-mediated ADCC against Panc-1, a human pancreatic cancer cell line with low HER2 expression, in vitro. Supplementation of anti-CD137 mAb could improve trastuzumab-mediated ADCC against Panc-1 which was insufficient without this stimulating antibody. The ADCC differed in individual cells, and this was related to the expression of CD137 on the surface of NK cells after trastuzumab stimulation in association with the Fcγ-RIIIA polymorphism. NK cells with Fcγ-RIIIA-VV/VF showed high levels of ADCC against Panc-1, but those with Fcγ-RIIIA-FF did not show optimal ADCC. In addition, trastuzumab-mediated ADCC against the human pancreatic cancer cell line Capan-1 with high HER2 expression was generally high and not affected by the Fcγ-RIIIA polymorphism. These results demonstrated that in Fcγ-RIIIA-VV/VF-carrying healthy individuals, trastuzumab plus αCD137 mAb could induce effective ADCC against HER2-low-expressing pancreatic cancer cell lines, and that such an approach may result in similar findings in patients with pancreatic cancer.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunity, Cellular/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Polymorphism, Genetic
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Trastuzumab/pharmacology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
- Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
Affiliation(s)
- Takushi Masu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Masanori Atsukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
- * E-mail:
| | - Katsuhisa Nakatsuka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Masumi Shimizu
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Daishu Miura
- Division of Breast and Thyroid Surgery, Toranomon Hospital, Tokyo, Japan
| | - Taeang Arai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Hirotomo Harimoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Chisa Kondo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Keiko Kaneko
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Seiji Futagami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Chiaki Kawamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Hidemi Takahashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Katsuhiko Iwakiri
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
194
|
Humphries MP, Hynes S, Bingham V, Cougot D, James J, Patel-Socha F, Parkes EE, Blayney JK, O'Rorke MA, Irwin GW, McArt DG, Kennedy RD, Mullan PB, McQuaid S, Salto-Tellez M, Buckley NE. Automated Tumour Recognition and Digital Pathology Scoring Unravels New Role for PD-L1 in Predicting Good Outcome in ER-/HER2+ Breast Cancer. JOURNAL OF ONCOLOGY 2018; 2018:2937012. [PMID: 30651729 PMCID: PMC6311859 DOI: 10.1155/2018/2937012] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/30/2018] [Accepted: 11/15/2018] [Indexed: 12/31/2022]
Abstract
The role of PD-L1 as a prognostic and predictive biomarker is an area of great interest. However, there is a lack of consensus on how to deliver PD-L1 as a clinical biomarker. At the heart of this conundrum is the subjective scoring of PD-L1 IHC in most studies to date. Current standard scoring systems involve separation of epithelial and inflammatory cells and find clinical significance in different percentages of expression, e.g., above or below 1%. Clearly, an objective, reproducible and accurate approach to PD-L1 scoring would bring a degree of necessary consistency to this landscape. Using a systematic comparison of technologies and the application of QuPath, a digital pathology platform, we show that high PD-L1 expression is associated with improved clinical outcome in Triple Negative breast cancer in the context of standard of care (SoC) chemotherapy, consistent with previous findings. In addition, we demonstrate for the first time that high PD-L1 expression is also associated with better outcome in ER- disease as a whole including HER2+ breast cancer. We demonstrate the influence of antibody choice on quantification and clinical impact with the Ventana antibody (SP142) providing the most robust assay in our hands. Through sampling different regions of the tumour, we show that tumour rich regions display the greatest range of PD-L1 expression and this has the most clinical significance compared to stroma and lymphoid rich areas. Furthermore, we observe that both inflammatory and epithelial PD-L1 expression are associated with improved survival in the context of chemotherapy. Moreover, as seen with PD-L1 inhibitor studies, a low threshold of PD-L1 expression stratifies patient outcome. This emphasises the importance of using digital pathology and precise biomarker quantitation to achieve accurate and reproducible scores that can discriminate low PD-L1 expression.
Collapse
Affiliation(s)
- Matthew P. Humphries
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Sean Hynes
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Victoria Bingham
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Delphine Cougot
- Horizon Discovery Ltd, 8100 Cambridge Research Park, Waterbeach, Cambridge, CB25 9TL, UK
| | - Jacqueline James
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Farah Patel-Socha
- Horizon Discovery Ltd, 8100 Cambridge Research Park, Waterbeach, Cambridge, CB25 9TL, UK
| | - Eileen E. Parkes
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Jaine K. Blayney
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Michael A. O'Rorke
- College of Public Health, The University of Iowa, Iowa City, IA 52242, USA
| | - Gareth W. Irwin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Darragh G. McArt
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Richard D. Kennedy
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Paul B. Mullan
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Stephen McQuaid
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Manuel Salto-Tellez
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Niamh E. Buckley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| |
Collapse
|
195
|
Lee S, Cohen DJ. Pharmacotherapy for metastatic esophageal cancer: where do we need to improve? Expert Opin Pharmacother 2018; 20:357-366. [PMID: 30526127 DOI: 10.1080/14656566.2018.1551881] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Esophageal cancer is a heterogeneous cancer comprised of differing cells of origin, molecular changes, and immune microenvironments. To date, most advances have been made in chemotherapy regimens where a one-size-fits-all approach is used. As a result, there remains a lack of tailored treatment options for such a heterogeneous cancer. This paper highlights the current standard of care treatment options as well as active areas of clinical research. AREAS COVERED The authors review the key trials that have led to current standard of care treatment including pivotal chemotherapy and targeted therapy trials. The authors then discuss the current approved uses and future directions for immunotherapy. EXPERT OPINION Current treatment options lack tailored treatment strategies based on the tumor's biology. To date, approved targeted approaches only include HER2-directed and anti-VEGFR2 therapies. Furthermore, while immunotherapy treatment response is often durable, few clear predictive biomarkers for response have been identified. Future research should focus on characterizing additional molecular targets for therapeutic intervention and predictive biomarkers for immunotherapy, as well as combination approaches of immunotherapy with other therapeutic modalities to increase response rate. Ultimately, the field should strive to develop personalized treatment options based on a tumor's molecular profile, microenvironment, and neo-antigen expression.
Collapse
Affiliation(s)
- Samuel Lee
- a Department of Medicine , Perlmutter Cancer Center, NYU Langone Medical Center , New York , NY , USA
| | - Deirdre J Cohen
- a Department of Medicine , Perlmutter Cancer Center, NYU Langone Medical Center , New York , NY , USA
| |
Collapse
|
196
|
Muntasell A, Rojo F, Servitja S, Rubio-Perez C, Cabo M, Tamborero D, Costa-García M, Martínez-Garcia M, Menéndez S, Vazquez I, Lluch A, Gonzalez-Perez A, Rovira A, López-Botet M, Albanell J. NK Cell Infiltrates and HLA Class I Expression in Primary HER2 + Breast Cancer Predict and Uncouple Pathological Response and Disease-free Survival. Clin Cancer Res 2018; 25:1535-1545. [PMID: 30523021 DOI: 10.1158/1078-0432.ccr-18-2365] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/17/2018] [Accepted: 11/30/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE We investigated the value of tumor-infiltrating NK (TI-NK) cells and HLA class I tumor expression as biomarkers of response to neoadjuvant anti-HER2 antibody-based treatment in breast cancer. EXPERIMENTAL DESIGN TI-NK cells and HLA-I were determined by IHC in pretreatment tumor biopsies from two cohorts of patients with HER2-positive breast cancer [discovery cohort (n = 42) and validation cohort (n = 71)]. Tumor-infiltrating lymphocytes (TIL) were scored according to international guidelines. Biomarker association with pathologic complete response (pCR) and disease-free survival (DFS) was adjusted for prognostic factors. Gene set variation analysis was used for determining immune cell populations concomitant to NK-cell enrichment in HER2-positive tumors from the Cancer Genome Atlas (n = 190). RESULTS TI-NK cells were significantly associated with pCR in the discovery cohort as well as in the validation cohort (P < 0.0001), independently of clinicopathologic factors. A ≥3 TI-NK cells/50x high-power field (HPF) cutoff predicted pCR in the discovery and validation cohort [OR, 188 (11-3154); OR, 19.5 (5.3-71.8)]. Presence of TI-NK cells associated with prolonged DFS in both patient cohorts [HR, 0.07 (0.01-0.6); P = 0.01; HR, 0.3 (0.08-1.3); P = 0.1]. NK-, activated dendritic- and CD8 T-cell gene expression signatures positively correlated in HER2-positive tumors, supporting the value of NK cells as surrogates of effective antitumor immunity. Stratification of patients by tumor HLA-I expression identified patients with low and high relapse risk independently of pCR. CONCLUSIONS This study identifies baseline TI-NK cells as an independent biomarker with great predictive value for pCR to anti-HER2 antibody-based treatment and points to the complementary value of tumor HLA-I status for defining patient prognosis independently of pCR.
Collapse
Affiliation(s)
- Aura Muntasell
- Immunity and Infection Lab, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.
| | - Federico Rojo
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Pathology, IIS 'Fundación Jiménez Diaz', Madrid, Spain
| | - Sonia Servitja
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Carlota Rubio-Perez
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mariona Cabo
- Immunity and Infection Lab, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - David Tamborero
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Oncology Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - María Martínez-Garcia
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Sílvia Menéndez
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain
| | - Ivonne Vazquez
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Ana Lluch
- Department of Oncology, Hospital Clinico de Valencia-CIBERONC, Valencia, Spain.,Universitat de Valencia, Valencia, Spain
| | - Abel Gonzalez-Perez
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - Miguel López-Botet
- Immunity and Infection Lab, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain.,Pompeu Fabra University, Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Institut Hospital del Mar d'Investigacions Mèdiques), Barcelona, Spain. .,Department of Medical Oncology, Hospital del Mar-CIBERONC, Barcelona, Spain.,Pompeu Fabra University, Barcelona, Spain
| |
Collapse
|
197
|
Triulzi T, Regondi V, De Cecco L, Cappelletti MR, Di Modica M, Paolini B, Lollini PL, Di Cosimo S, Sfondrini L, Generali D, Tagliabue E. Early immune modulation by single-agent trastuzumab as a marker of trastuzumab benefit. Br J Cancer 2018; 119:1487-1494. [PMID: 30478407 PMCID: PMC6288086 DOI: 10.1038/s41416-018-0318-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Optimising the selection of HER2-targeted regimens by identifying subsets of HER2-positive breast cancer (BC) patients who need more or less therapy remains challenging. We analysed BC samples before and after treatment with 1 cycle of trastuzumab according to the response to trastuzumab. METHODS Gene expression profiles of pre- and post-treatment tumour samples from 17 HER2-positive BC patients were analysed on the Illumina platform. Tumour-associated immune pathways and blood counts were analysed with regard to the response to trastuzumab. HER2-positive murine models with differential responses to trastuzumab were used to reproduce and better characterise these data. RESULTS Patients who responded to single-agent trastuzumab had basal tumour biopsies that were enriched in immune pathways, particularly the MHC-II metagene. One cycle of trastuzumab modulated the expression levels of MHC-II genes, which increased in patients who had a complete response on treatment with trastuzumab and chemotherapy. Trastuzumab increased the MHC-II-positive cell population, primarily macrophages, only in the tumour microenvironment of responsive mice. In patients who benefited from complete trastuzumab therapy and in mice that harboured responsive tumours circulating neutrophil levels declined, but this cell subset rose in nonresponsive tumours. CONCLUSIONS Short treatment with trastuzumab induces local and systemic immunomodulation that is associated with clinical outcomes.
Collapse
Affiliation(s)
- Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Viola Regondi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Loris De Cecco
- Platform of Integrated Biology, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Maria Rosa Cappelletti
- U.O. Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, Azienda Socio-Sanitaria Territoriale di Cremona, Cremona, Italy
| | - Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Biagio Paolini
- Anatomic Pathology A Unit, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Pier Luigi Lollini
- Laboratory of Immunology and Biology of Metastases, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Serena Di Cosimo
- Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milano, Italy
| | - Daniele Generali
- U.O. Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, Azienda Socio-Sanitaria Territoriale di Cremona, Cremona, Italy
- Dipartimento Universitario Clinico di Scienze Mediche, Chirurgiche e della Salute, Università degli Studi di Trieste, Trieste, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy.
| |
Collapse
|
198
|
Christmas BJ, Rafie CI, Hopkins AC, Scott BA, Ma HS, Cruz KA, Woolman S, Armstrong TD, Connolly RM, Azad NA, Jaffee EM, Roussos Torres ET. Entinostat Converts Immune-Resistant Breast and Pancreatic Cancers into Checkpoint-Responsive Tumors by Reprogramming Tumor-Infiltrating MDSCs. Cancer Immunol Res 2018; 6:1561-1577. [PMID: 30341213 PMCID: PMC6279584 DOI: 10.1158/2326-6066.cir-18-0070] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/08/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022]
Abstract
Immune-checkpoint inhibition (ICI) has revolutionized treatment in cancers that are naturally immunogenic by enabling infiltration of T cells into the tumor microenvironment (TME) and promoting cytotoxic signaling pathways. Tumors possessing complex immunosuppressive TMEs such as breast and pancreatic cancers present unique therapeutic obstacles as response rates to ICI remain low. Such tumors often recruit myeloid-derived suppressor cells (MDSCs), whose functioning prohibits both T-cell activation and infiltration. We attempted to sensitize these tumors to ICI using epigenetic modulation to target MDSC trafficking and function to foster a less immunosuppressive TME. We showed that combining a histone deacetylase inhibitor, entinostat (ENT), with anti-PD-1, anti-CTLA-4, or both significantly improved tumor-free survival in both the HER2/neu transgenic breast cancer and the Panc02 metastatic pancreatic cancer mouse models. Using flow cytometry, gene-expression profiling, and ex vivo functional assays, we characterized populations of tumor-infiltrating lymphocytes (TILs) and MDSCs, as well as their functional capabilities. We showed that addition of ENT to checkpoint inhibition led to significantly decreased suppression by granulocytic MDSCs in the TME of both tumor types. We also demonstrated an increase in activated granzyme-B-producing CD8+ T effector cells in mice treated with combination therapy. Gene-expression profiling of both MDSCs and TILs identified significant changes in immune-related pathways. In summary, addition of ENT to ICI significantly altered infiltration and function of innate immune cells, allowing for a more robust adaptive immune response. These findings provide a rationale for combination therapy in patients with immune-resistant tumors, including breast and pancreatic cancers.
Collapse
MESH Headings
- Animals
- Female
- Male
- Antineoplastic Agents/pharmacology
- Benzamides/pharmacology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/mortality
- CTLA-4 Antigen/antagonists & inhibitors
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/pathology
- Mice, Inbred C57BL
- Mice, Transgenic
- Myeloid-Derived Suppressor Cells/drug effects
- Myeloid-Derived Suppressor Cells/immunology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Pyridines/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
- Mice
Collapse
Affiliation(s)
- Brian J Christmas
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine I Rafie
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander C Hopkins
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Blake A Scott
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hayley S Ma
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kayla A Cruz
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Skylar Woolman
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Todd D Armstrong
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Roisin M Connolly
- Department of Oncology, and the Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nilo A Azad
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, and the Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, and the Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Evanthia T Roussos Torres
- Viragh Center for Pancreatic Clinical Research and Care, Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, and the Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
199
|
|
200
|
Benonisson H, Sow HS, Breukel C, Claassens J, Brouwers C, Linssen MM, Fransen MF, Sluijter M, Ossendorp F, van Hall T, Verbeek JS. High FcγR Expression on Intratumoral Macrophages Enhances Tumor-Targeting Antibody Therapy. THE JOURNAL OF IMMUNOLOGY 2018; 201:3741-3749. [PMID: 30397036 DOI: 10.4049/jimmunol.1800700] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022]
Abstract
Therapy with tumor-specific Abs is common in the clinic but has limited success against solid malignancies. We aimed at improving the efficacy of this therapy by combining a tumor-specific Ab with immune-activating compounds. In this study, we demonstrate in the aggressive B16F10 mouse melanoma model that concomitant application of the anti-TRP1 Ab (clone TA99) with TLR3-7/8 or -9 ligands, and IL-2 strongly enhanced tumor control in a therapeutic setting. Depletion of NK cells, macrophages, or CD8+ T cells all mitigated the therapeutic response, showing a coordinated immune rejection by innate and adaptive immune cells. FcγRs were essential for the therapeutic effect, with a dominant role for FcγRI and a minor role for FcγRIII and FcγRIV. FcγR expression on NK cells and granulocytes was dispensable, indicating that other tumoricidal functions of NK cells were involved and implicating that FcγRI, -III, and -IV exerted their activity on macrophages. Indeed, F4/80+Ly-6C+ inflammatory macrophages in the tumor microenvironment displayed high levels of these receptors. Whereas administration of the anti-TRP1 Ab alone reduced the frequency of these macrophages, the combination with a TLR agonist retained these cells in the tumor microenvironment. Thus, the addition of innate stimulatory compounds, such as TLR ligands, to tumor-specific Ab therapy could greatly enhance its efficacy in solid cancers via optimal exploitation of FcγRs.
Collapse
Affiliation(s)
- Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Heng Sheng Sow
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Cor Breukel
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jill Claassens
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Conny Brouwers
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Margot M Linssen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands;
| |
Collapse
|