1
|
Owens K, Rahman A, Bozic I. Spatiotemporal dynamics of tumor - CAR T-cell interaction following local administration in solid cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610392. [PMID: 39257746 PMCID: PMC11384001 DOI: 10.1101/2024.08.29.610392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The success of chimeric antigen receptor (CAR) T-cell therapy in treating hematologic malignancies has generated widespread interest in translating this technology to solid cancers. However, issues like tumor infiltration, the immunosuppressive tumor microenvironment, and tumor heterogeneity limit its efficacy in the solid tumor setting. Recent experimental and clinical studies propose local administration directly into the tumor or at the tumor site to increase CAR T-cell infiltration and improve treatment outcomes. Characteristics of the types of solid tumors that may be the most receptive to this treatment approach remain unclear. In this work, we develop a spatiotemporal model for CAR T-cell treatment of solid tumors, and use numerical simulations to compare the effect of introducing CAR T cells via intratumoral injection versus intracavitary administration in diverse cancer types. We demonstrate that the model can recapitulate tumor and CAR T-cell data from imaging studies of local administration of CAR T cells in mouse models. Our results suggest that locally administered CAR T cells will be most successful against slowly proliferating, highly diffusive tumors, which have the lowest average tumor cell density. These findings affirm the clinical observation that CAR T cells will not perform equally across different types of solid tumors, and suggest that measuring tumor density may be helpful when considering the feasibility of CAR T-cell therapy and planning dosages for a particular patient. We additionally find that local delivery of CAR T cells can result in deep tumor responses, provided that the initial CAR T-cell dose does not contain a significant fraction of exhausted cells.
Collapse
Affiliation(s)
- Katherine Owens
- Department of Applied Mathematics, University of Washington, Seattle WA
- Fred Hutchinson Cancer Center, Seattle WA
| | - Aminur Rahman
- Fred Hutchinson Cancer Center, Seattle WA
- Artificial Intelligence Institute in Dynamic Systems, University of Washington, Seattle WA
| | - Ivana Bozic
- Department of Applied Mathematics, University of Washington, Seattle WA
- Fred Hutchinson Cancer Center, Seattle WA
| |
Collapse
|
2
|
Pandey P, Chaudhary R, Tripathi D, Lavudi K, Dua K, Weinfeld M, Lavasanifar A, Rajinikanth PS. Personalized treatment approach for HER2-positive metastatic breast cancer. Med Oncol 2024; 41:252. [PMID: 39320608 DOI: 10.1007/s12032-024-02504-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Breast cancer (BC) is a leading global concern for women, with 30% being HER2-positive cases linked to poorer outcomes. Targeted therapies like trastuzumab deruxtecan (T-DXd), trastuzumab, pertuzumab, and T-DM1 have revolutionized HER2-positive metastatic breast cancer (MBC) treatment. Although these therapies have improved MBC management and patient outcomes, resistance can develop, reducing effectiveness. Personalized strategies based on tumor characteristics offer hope for better responses and longer outcomes. This review outlines insights into MBC patients responding well to anti-HER2 treatments, even across multiple treatment regimen. Recent immunotherapy, locoregional therapy, and liquid biopsy breakthroughs are covered, suggesting ways to increase long-term responders. Personalized approaches have boosted HER2-positive MBC outcomes, and ongoing research is crucial to uncover new treatments and biomarkers, potentially elevating long-term response rates and prognoses. This may aid in providing new direction to breast cancer clinics.
Collapse
Affiliation(s)
- Prashant Pandey
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India
| | - Devika Tripathi
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), Kanpur, India
| | - Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Michael Weinfeld
- Cross Cancer Institute and Department of Oncology, University of Alberta, Edmonton, AB, T6G 1Z2, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Department of Chemical and Material Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
3
|
Heidarnejad K, Nooreddin Faraji S, Mahfoozi S, Ghasemi Z, Sadat Dashti F, Asadi M, Ramezani A. Breast cancer immunotherapy using scFv antibody-based approaches, a systematic review. Hum Immunol 2024; 85:111090. [PMID: 39214066 DOI: 10.1016/j.humimm.2024.111090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer is considered as the most common malignancy in women and the second leading cause of death related to cancer. Recombinant DNA technologies accelerated the development of antibody-based cancer therapy, which is effective in a broad range of cancers. The objective of the present study was to perform a systematic review on breast cancer immunotherapy using single-chain fragment variable (scFv) antibody formats. Searches were performed up to March 2023 using PubMed, Scopus, and Web of Science (ISI) databases. Three reviewers independently assessed study eligibility, data extraction, and evaluated the methodological quality of included primary studies. Different immunotherapy approaches have been identified and the most common approaches were scFv-conjugates, followed by simple scFvs and chimeric antigen receptor (CAR) therapy, respectively. Among breast cancer antigens, HER superfamily, CD family, and EpCAM were applied as the most important breast cancer immunotherapy targets. The present study shed more lights on scFv-based breast cancer immunotherapy approaches.
Collapse
Affiliation(s)
- Kamran Heidarnejad
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Nooreddin Faraji
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Shirin Mahfoozi
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ghasemi
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fateme Sadat Dashti
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maryam Asadi
- School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Holtermann A, Gislon M, Angele M, Subklewe M, von Bergwelt-Baildon M, Lauber K, Kobold S. Prospects of Synergy: Local Interventions and CAR T Cell Therapy in Solid Tumors. BioDrugs 2024; 38:611-637. [PMID: 39080180 PMCID: PMC11358237 DOI: 10.1007/s40259-024-00669-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2024] [Indexed: 08/30/2024]
Abstract
Chimeric antigen receptor T cell therapy has been established in the treatment of various B cell malignancies. However, translating this therapeutic effect to treat solid tumors has been challenging because of their inter-tumoral as well as intratumoral heterogeneity and immunosuppressive microenvironment. Local interventions, such as surgery, radiotherapy, local ablation, and locoregional drug delivery, can enhance chimeric antigen receptor T cell therapy in solid tumors by improving tumor infiltration and reducing systemic toxicities. Additionally, ablation and radiotherapy have proven to (re-)activate systemic immune responses via abscopal effects and reprogram the tumor microenvironment on a physical, cellular, and chemical level. This review highlights the potential synergy of the combined approaches to overcome barriers of chimeric antigen receptor T cell therapy and summarizes recent studies that may pave the way for new treatment regimens.
Collapse
Affiliation(s)
- Anne Holtermann
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Mila Gislon
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health Neuherberg, Munich, Germany.
| |
Collapse
|
5
|
Erler P, Kurcon T, Cho H, Skinner J, Dixon C, Grudman S, Rozlan S, Dessez E, Mumford B, Jo S, Boyne A, Juillerat A, Duchateau P, Poirot L, Aranda-Orgilles B. Multi-armored allogeneic MUC1 CAR T cells enhance efficacy and safety in triple-negative breast cancer. SCIENCE ADVANCES 2024; 10:eadn9857. [PMID: 39213364 PMCID: PMC11364110 DOI: 10.1126/sciadv.adn9857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
Solid tumors, such as triple-negative breast cancer (TNBC), are biologically complex due to cellular heterogeneity, lack of tumor-specific antigens, and an immunosuppressive tumor microenvironment (TME). These challenges restrain chimeric antigen receptor (CAR) T cell efficacy, underlining the importance of armoring. In solid cancers, a localized tumor mass allows alternative administration routes, such as intratumoral delivery with the potential to improve efficacy and safety but may compromise metastatic-site treatment. Using a multi-layered CAR T cell engineering strategy that allowed a synergy between attributes, we show enhanced cytotoxic activity of MUC1 CAR T cells armored with PD1KO, tumor-specific interleukin-12 release, and TGFBR2KO attributes catered towards the TNBC TME. Intratumoral treatment effectively reduced distant tumors, suggesting retention of antigen-recognition benefits at metastatic sites. Overall, we provide preclinical evidence of armored non-alloreactive MUC1 CAR T cells greatly reducing high TNBC tumor burden in a TGFB1- and PD-L1-rich TME both at local and distant sites while preserving safety.
Collapse
Affiliation(s)
| | | | - Hana Cho
- Cellectis Inc., New York, NY, USA
| | | | | | | | | | | | | | - Sumin Jo
- Cellectis Inc., New York, NY, USA
| | | | | | | | | | | |
Collapse
|
6
|
Nipper AJ, Warren EAK, Liao KS, Liu HC, Michikawa C, Porter CE, Wells GA, Villanueva M, Brasil da Costa FH, Veeramachaneni R, Villanueva H, Suzuki M, Sikora AG. Chick Embryo Chorioallantoic Membrane as a Platform for Assessing the In Vivo Efficacy of Chimeric Antigen Receptor T-cell Therapy in Solid Tumors. Immunohorizons 2024; 8:598-605. [PMID: 39225630 PMCID: PMC11374747 DOI: 10.4049/immunohorizons.2400059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
The fertilized chicken egg chorioallantoic membrane (CAM), a highly vascularized membrane nourishing the developing embryo, also supports rapid growth of three-dimensional vascularized tumors from engrafted cells and tumor explants. Because murine xenograft models suffer limitations of time, cost, and scalability, we propose CAM tumors as a rapid, efficient screening tool for assessing anti-tumor efficacy of chimeric Ag receptor (CAR) T cells against solid tumors. We tested the efficacy of human epidermal growth factor receptor 2 (HER2)-specific CAR T cells against luminescent, HER2-expressing (FaDu, SCC-47) or HER2-negative (MDA-MB-468) CAM-engrafted tumors. Three days after tumor engraftment, HER2-specific CAR T cells were applied to tumors grown on the CAM. Four days post-CAR T cell treatment, HER2-expressing FaDu and SCC-47 tumors treated with CAR T showed reduced viable cancer cells as assessed by luciferase activity. This reduction in viable tumor cells was confirmed by histology, with lower Ki-67 staining observed in CAR T cell-treated tumors relative to T cell-treated controls. Persistence of CAR T in CAM and tumor tissue 4 days post-treatment was confirmed by CD3 staining. Altogether, our findings support further development of the chick CAM as an in vivo system for rapid, scalable screening of CAR T cell efficacy against human solid tumors.
Collapse
Affiliation(s)
- Allison J. Nipper
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Emilie A. K. Warren
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX
| | - Kershena S. Liao
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX
| | - Hsuan-Chen Liu
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX
| | - Chieko Michikawa
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Caroline E. Porter
- Department of Family and Community Medicine, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | | | - Mariana Villanueva
- Department of Family and Community Medicine, Baylor College of Medicine, Houston, TX
| | | | - Ratna Veeramachaneni
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Hugo Villanueva
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX
| | - Masataka Suzuki
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
- Texas Children’s Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Andrew G. Sikora
- Department of Head and Neck Surgery, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
7
|
Mantooth SM, Abdou Y, Saez-Ibañez AR, Upadhaya S, Zaharoff DA. Intratumoral delivery of immunotherapy to treat breast cancer: current development in clinical and preclinical studies. Front Immunol 2024; 15:1385484. [PMID: 38803496 PMCID: PMC11128577 DOI: 10.3389/fimmu.2024.1385484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Breast cancer poses one of the largest threats to women's health. Treatment continues to improve for all the subtypes of breast cancer, but some subtypes, such as triple negative breast cancer, still present a significant treatment challenge. Additionally, metastasis and local recurrence are two prevalent problems in breast cancer treatment. A newer type of therapy, immunotherapy, may offer alternatives to traditional treatments for difficult-to-treat subtypes. Immunotherapy engages the host's immune system to eradicate disease, with the potential to induce long-lasting, durable responses. However, systemic immunotherapy is only approved in a limited number of indications, and it benefits only a minority of patients. Furthermore, immune related toxicities following systemic administration of potent immunomodulators limit dosing and, consequently, efficacy. To address these safety considerations and improve treatment efficacy, interest in local delivery at the site of the tumor has increased. Numerous intratumorally delivered immunotherapeutics have been and are being explored clinically and preclinically, including monoclonal antibodies, cellular therapies, viruses, nucleic acids, cytokines, innate immune agonists, and bacteria. This review summarizes the current and past intratumoral immunotherapy clinical landscape in breast cancer as well as current progress that has been made in preclinical studies, with a focus on delivery parameters and considerations.
Collapse
Affiliation(s)
- Siena M. Mantooth
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
| | - Yara Abdou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | - David A. Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
8
|
Nagy L, Mezősi-Csaplár M, Rebenku I, Vereb G, Szöőr Á. Universal CAR T cells targeted to HER2 with a biotin-trastuzumab soluble linker penetrate spheroids and large tumor xenografts that are inherently resistant to trastuzumab mediated ADCC. Front Immunol 2024; 15:1365172. [PMID: 38562932 PMCID: PMC10982377 DOI: 10.3389/fimmu.2024.1365172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
CAR T cell therapies face challenges in combating solid tumors due to their single-target approach, which becomes ineffective if the targeted antigen is absent or lost. Universal CAR T cells (UniCAR Ts) provide a promising solution by utilizing molecular tags (linkers), such as biotin conjugated to monoclonal antibodies, enabling them to target a variety of tumor antigens. Recently, we showed that conventional CAR T cells could penetrate the extracellular matrix (ECM) of ADCC-resistant tumors, which forms a barrier to therapeutic antibodies. This finding led us to investigate whether UniCAR T cells, targeted by soluble antibody-derived linkers, could similarly tackle ADCC-resistant tumors where ECM restricts antibody penetration. We engineered UniCAR T cells by incorporating a biotin-binding monomeric streptavidin 2 (mSA2) domain for targeting HER2 via biotinylated trastuzumab (BT). The activation and cytotoxicity of UniCAR T cells in the presence or absence of BT were evaluated in conventional immunoassays. A 3D spheroid coculture was set up to test the capability of UniCAR Ts to access ECM-masked HER2+ cells. For in vivo analysis, we utilized a HER2+ xenograft model in which intravenously administered UniCAR T cells were supplemented with intraperitoneal BT treatments. In vitro, BT-guided UniCAR T cells showed effective activation and distinct anti-tumor response. Upon target recognition, IFNγ secretion correlated with BT concentration. In the presence of BT, UniCAR T cells effectively penetrated HER2+ spheroids and induced cell death in their core regions. In vivo, upon intravenous administration of UniCAR Ts, circulating BT linkers immediately engaged the mSA2 domain and directed effector cells to the HER2+ tumors. However, these co-treated mice died early, possibly due to the lung infiltration of UniCAR T cells that could recognize both native biotin and HER2. Our results suggest that UniCAR T cells guided with soluble linkers present a viable alternative to conventional CAR T cells, especially for patients resistant to antibody therapy and those with solid tumors exhibiting high antigenic variability. Critical to their success, however, is the choice of an appropriate binding domain for the CAR and the corresponding soluble linker, ensuring both efficacy and safety in therapeutic applications.
Collapse
Affiliation(s)
- Lőrinc Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Marianna Mezősi-Csaplár
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Rebenku
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
9
|
Chamorro DF, Somes LK, Hoyos V. Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential. Cancers (Basel) 2023; 16:124. [PMID: 38201551 PMCID: PMC10778447 DOI: 10.3390/cancers16010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Breast cancer remains a significant health challenge, and novel treatment approaches are critically needed. This review presents an in-depth analysis of engineered adoptive T-cell therapies (E-ACTs), an innovative frontier in cancer immunotherapy, focusing on their application in breast cancer. We explore the evolving landscape of chimeric antigen receptor (CAR) and T-cell receptor (TCR) T-cell therapies, highlighting their potential and challenges in targeting breast cancer. The review addresses key obstacles such as target antigen selection, the complex breast cancer tumor microenvironment, and the persistence of engineered T-cells. We discuss the advances in overcoming these barriers, including strategies to enhance T-cell efficacy. Finally, our comprehensive analysis of the current clinical trials in this area provides insights into the future possibilities and directions of E-ACTs in breast cancer treatment.
Collapse
Affiliation(s)
- Diego F. Chamorro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Lauren K. Somes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
Cao Y, Efetov SK, He M, Fu Y, Beeraka NM, Zhang J, Zhang X, Bannimath N, Chen K. Updated Clinical Perspectives and Challenges of Chimeric Antigen Receptor-T Cell Therapy in Colorectal Cancer and Invasive Breast Cancer. Arch Immunol Ther Exp (Warsz) 2023; 71:19. [DOI: https:/doi.org/10.1007/s00005-023-00684-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/28/2023] [Indexed: 09/20/2024]
|
11
|
Shyr CR, Liu LC, Chien HS, Huang CP. Immunotherapeutic Agents for Intratumoral Immunotherapy. Vaccines (Basel) 2023; 11:1717. [PMID: 38006049 PMCID: PMC10674963 DOI: 10.3390/vaccines11111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/22/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Immunotherapy using systemic immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cells has revolutionized cancer treatment, but it only benefits a subset of patients. Systemic immunotherapies cause severe autoimmune toxicities and cytokine storms. Immune-related adverse events (irAEs) plus the immunosuppressive tumor microenvironment (TME) have been linked to the inefficacy of systemic immunotherapy. Intratumoral immunotherapy that increases immunotherapeutic agent bioavailability inside tumors could enhance the efficacy of immunotherapies and reduce systemic toxicities. In preclinical and clinical studies, intratumoral administration of immunostimulatory agents from small molecules to xenogeneic cells has demonstrated antitumor effects not only on the injected tumors but also against noninjected lesions. Herein, we review and discuss the results of these approaches in preclinical models and clinical trials to build the landscape of intratumoral immunotherapeutic agents and we describe how they stimulate the body's immune system to trigger antitumor immunity as well as the challenges in clinical practice. Systemic and intratumoral combination immunotherapy would make the best use of the body's immune system to treat cancers. Combining precision medicine and immunotherapy in cancer treatment would treat both the mutated targets in tumors and the weakened body's immune system simultaneously, exerting maximum effects of the medical intervention.
Collapse
Affiliation(s)
- Chih-Rong Shyr
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404328, Taiwan; (C.-R.S.); (H.-S.C.)
- eXCELL Biotherapeutics Inc., Taichung 404328, Taiwan
| | - Lang-Chi Liu
- Department of Medicine, Department of Surgery, College of Medicine, China Medical University and Hospital, Taichung 404328, Taiwan;
| | - Hui-Shan Chien
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404328, Taiwan; (C.-R.S.); (H.-S.C.)
| | - Chi-Ping Huang
- Department of Medicine, Urology Division, China Medical University and Hospital, Taichung 404328, Taiwan
| |
Collapse
|
12
|
Cao Y, Efetov SK, He M, Fu Y, Beeraka NM, Zhang J, Zhang X, Bannimath N, Chen K. Updated Clinical Perspectives and Challenges of Chimeric Antigen Receptor-T Cell Therapy in Colorectal Cancer and Invasive Breast Cancer. Arch Immunol Ther Exp (Warsz) 2023; 71:19. [PMID: 37566162 DOI: 10.1007/s00005-023-00684-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 06/28/2023] [Indexed: 08/12/2023]
Abstract
In recent years, the incidence of colorectal cancer (CRC) and breast cancer (BC) has increased worldwide and caused a higher mortality rate due to the lack of selective anti-tumor therapies. Current chemotherapies and surgical interventions are significantly preferred modalities to treat CRC or BC in advanced stages but the prognosis for patients with advanced CRC and BC remains dismal. The immunotherapy technique of chimeric antigen receptor (CAR)-T cells has resulted in significant clinical outcomes when treating hematologic malignancies. The novel CAR-T therapy target antigens include GUCY2C, CLEC14A, CD26, TEM8/ANTXR1, PDPN, PTK7, PODXL, CD44, CD19, CD20, CD22, BCMA, GD2, Mesothelin, TAG-72, CEA, EGFR, B7H3, HER2, IL13Ra2, MUC1, EpCAM, PSMA, PSCA, NKG2D. The significant aim of this review is to explore the recently updated information pertinent to several novel targets of CAR-T for CRC, and BC. We vividly described the challenges of CAR-T therapies when treating CRC or BC. The immunosuppressive microenvironment of solid tumors, the shortage of tumor-specific antigens, and post-treatment side effects are the major hindrances to promoting the development of CAR-T cells. Several clinical trials related to CAR-T immunotherapy against CRC or BC have already been in progress. This review benefits academicians, clinicians, and clinical oncologists to explore more about the novel CAR-T targets and overcome the challenges during this therapy.
Collapse
Affiliation(s)
- Yu Cao
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Sergey K Efetov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Mingze He
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Yu Fu
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Narasimha M Beeraka
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh, 515721, India
| | - Jin Zhang
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Xinliang Zhang
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Namitha Bannimath
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Kuo Chen
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, #1 Jianshedong Str., Zhengzhou, 450052, People's Republic of China.
| |
Collapse
|
13
|
Sailer CJ, Hong Y, Dahal A, Ryan AT, Mir S, Gerber SA, Reagan PM, Kim M. PD-1 Hi CAR-T cells provide superior protection against solid tumors. Front Immunol 2023; 14:1187850. [PMID: 37388744 PMCID: PMC10303811 DOI: 10.3389/fimmu.2023.1187850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising treatment option for several hematologic cancers. However, efforts to achieve the same level of therapeutic success in solid tumors have largely failed mainly due to CAR-T cell exhaustion and poor persistence at the tumor site. Although immunosuppression mediated by augmented programmed cell death protein-1 (PD-1) expression has been proposed to cause CAR-T cell hypofunction and limited clinical efficacy, little is known about the underlying mechanisms and immunological consequences of PD-1 expression on CAR-T cells. With flow cytometry analyses and in vitro and in vivo anti-cancer T cell function assays, we found that both manufactured murine and human CAR-T cell products displayed phenotypic signs of T cell exhaustion and heterogeneous expression levels of PD-1. Unexpectedly, PD-1high CAR-T cells outperformed PD-1low CAR-T cells in multiple T cell functions both in vitro and in vivo. Despite the achievement of superior persistence at the tumor site in vivo, adoptive transfer of PD-1high CAR-T cells alone failed to control tumor growth. Instead, a PD-1 blockade combination therapy significantly delayed tumor progression in mice infused with PD-1high CAR-T cells. Therefore, our data demonstrate that robust T cell activation during the ex vivo CAR-T cell manufacturing process generates a PD-1high CAR-T cell subset with improved persistence and enhanced anti-cancer functions. However, these cells may be vulnerable to the immunosuppressive microenvironment and require combination with PD-1 inhibition to maximize therapeutic functions in solid tumors.
Collapse
Affiliation(s)
- Cooper J. Sailer
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
| | - Yeonsun Hong
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Ankit Dahal
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Allison T. Ryan
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Sana Mir
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| | - Scott A. Gerber
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Patrick M. Reagan
- Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, United States
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
14
|
Yin L, Chen GL, Xiang Z, Liu YL, Li XY, Bi JW, Wang Q. Current progress in chimeric antigen receptor-modified T cells for the treatment of metastatic breast cancer. Biomed Pharmacother 2023; 162:114648. [PMID: 37023621 DOI: 10.1016/j.biopha.2023.114648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Breast cancer is the leading cancer in women. Around 20-30% breast cancer patients undergo invasion or metastasis after radical surgical resection and eventually die. Number of breast cancer patients show poor sensitivity toward treatments despite the advances in chemotherapy, endocrine therapy, and molecular targeted treatments. Therapeutic resistance and tumor recurrence or metastasis develop with the ongoing treatments. Conducive treatment strategies are thus required. Chimeric antigen receptor (CAR)-modified T-cell therapy has progressed as a part of tumor immunotherapy. However, CAR-T treatment has not been effective in solid tumors because of tumor microenvironment complexity, inhibitory effects of extracellular matrix, and lacking ideal tumor antigens. Herein, the prospects of CAR-T cell therapy for metastatic breast cancer are discussed, and the targets for CAR-T therapy in breast cancer (HER-2, C-MET, MSLN, CEA, MUC1, ROR1, EGFR) at clinical level are reviewed. Moreover, solutions are proposed for the challenges of breast cancer CAR-T therapy regarding off-target effects, heterogeneous antigen expression by tumor cells and immunosuppressive tumor microenvironment. Ideas for improving the therapeutics of CAR-T cell therapy in metastatic breast cancer are suggested.
Collapse
Affiliation(s)
- Li Yin
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China; Shandong University of Traditional Chinese Medicine, 250355 Jinan, China
| | - Gui-Lai Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Zhuo Xiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Yu-Lin Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China
| | - Xing-Yu Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, China
| | - Jing-Wang Bi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China.
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, 250023 Jinan, China; Oncology Department, Shandong Second Provincial General Hospital, 250023 Jinan, China; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 266003 Qingdao, China.
| |
Collapse
|
15
|
Xie L, Meng Z. Immunomodulatory effect of locoregional therapy in the tumor microenvironment. Mol Ther 2023; 31:951-969. [PMID: 36694462 PMCID: PMC10124087 DOI: 10.1016/j.ymthe.2023.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/15/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Cancer immunotherapy appears to be a promising treatment option; however, only a subset of patients with cancer responds favorably to treatment. Locoregional therapy initiates a local antitumor immune response by disrupting immunosuppressive components, releasing immunostimulatory damage-associated molecular patterns, recruiting immune effectors, and remodeling the tumor microenvironment. Many studies have shown that locoregional therapy can produce specific antitumor immunity alone; nevertheless, the effect is relatively weak and transient. Furthermore, increasing research efforts have explored the potential synergy between locoregional therapy and immunotherapy to enhance the long-term systemic antitumor immune effect and improve survival. Therefore, further research is needed into the immunomodulatory effects of locoregional therapy and immunotherapy to augment antitumor effects. This review article summarizes the key components of the tumor microenvironment, discusses the immunomodulatory role of locoregional therapy in the tumor microenvironment, and emphasizes the therapeutic potential of locoregional therapy in combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Lin Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Zhiqiang Meng
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China.
| |
Collapse
|
16
|
Mercogliano MF, Bruni S, Mauro FL, Schillaci R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15071987. [PMID: 37046648 PMCID: PMC10093019 DOI: 10.3390/cancers15071987] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of death. HER2 overexpression is found in approximately 20% of breast cancers and is associated with a poor prognosis and a shorter overall survival. Tratuzumab, a monoclonal antibody directed against the HER2 receptor, is the standard of care treatment. However, a third of the patients do not respond to therapy. Given the high rate of resistance, other HER2-targeted strategies have been developed, including monoclonal antibodies such as pertuzumab and margetuximab, trastuzumab-based antibody drug conjugates such as trastuzumab-emtansine (T-DM1) and trastuzumab-deruxtecan (T-DXd), and tyrosine kinase inhibitors like lapatinib and tucatinib, among others. Moreover, T-DXd has proven to be of use in the HER2-low subtype, which suggests that other HER2-targeted therapies could be successful in this recently defined new breast cancer subclassification. When patients progress to multiple strategies, there are several HER2-targeted therapies available; however, treatment options are limited, and the potential combination with other drugs, immune checkpoint inhibitors, CAR-T cells, CAR-NK, CAR-M, and vaccines is an interesting and appealing field that is still in development. In this review, we will discuss the highlights and pitfalls of the different HER2-targeted therapies and potential combinations to overcome metastatic disease and resistance to therapy.
Collapse
|
17
|
Altinoz MA, Ozpinar A, Hacker E, Ozpinar A. Combining locoregional CAR-T cells, autologous + allogeneic tumor lysate vaccination and levamisole in treatment of glioblastoma. Immunopharmacol Immunotoxicol 2022; 44:797-808. [PMID: 35670420 DOI: 10.1080/08923973.2022.2086136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain malignancy and harbors a microenvironment limiting immune cells activity. CAR-T cells are being tested in the treatment of cancers and there exist reports which demonstrate dramatic regression of multicentric GBMs following intrathecal treatment with CAR-T cells. In this article, a triple approach for immune treatment of GBM is proposed. First, GBM tumor specimens for each patient will be saved and cultured to obtain tumor lysates. Then, levamisole will be applied, which possesses immunostimulating, anti-glycolytic, and anti-angiogenic features. Following priming the immune system, GBM patients will be injected with lysates of their own tumor cells plus lysates from a GBM cell line, U251. After 3 months of this treatment, CAR-T cells (transduced with IL13Rα2-CAR) will be applied via intratumoral approach. As such, genetically-modified and native immunocytes may 'meet' in the vicinity of deeply-invading tumor cells and demonstrate greater efficacy via cell-cell interactions. By this, a self-propagating cyclic process - a cancer-immunity cycle - may be initiated to eradicate cancer cells.
Collapse
Affiliation(s)
- Meric A Altinoz
- Department of Biochemistry, Acibadem University, Istanbul, Turkey
| | - Alp Ozpinar
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily Hacker
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aysel Ozpinar
- Department of Biochemistry, Acibadem University, Istanbul, Turkey
| |
Collapse
|
18
|
Gámez-Chiachio M, Sarrió D, Moreno-Bueno G. Novel Therapies and Strategies to Overcome Resistance to Anti-HER2-Targeted Drugs. Cancers (Basel) 2022; 14:4543. [PMID: 36139701 PMCID: PMC9496705 DOI: 10.3390/cancers14184543] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The prognosis and quality of life of HER2 breast cancer patients have significantly improved due to the crucial clinical benefit of various anti-HER2 targeted therapies. However, HER2 tumors can possess or develop several resistance mechanisms to these treatments, thus leaving patients with a limited set of additional therapeutic options. Fortunately, to overcome this problem, in recent years, multiple different and complementary approaches have been developed (such as antibody-drug conjugates (ADCs)) that are in clinical or preclinical stages. In this review, we focus on emerging strategies other than on ADCs that are either aimed at directly target the HER2 receptor (i.e., novel tyrosine kinase inhibitors) or subsequent intracellular signaling (e.g., PI3K/AKT/mTOR, CDK4/6 inhibitors, etc.), as well as on innovative approaches designed to attack other potential tumor weaknesses (such as immunotherapy, autophagy blockade, or targeting of other genes within the HER2 amplicon). Moreover, relevant technical advances such as anti-HER2 nanotherapies and immunotoxins are also discussed. In brief, this review summarizes the impact of novel therapeutic approaches on current and future clinical management of aggressive HER2 breast tumors.
Collapse
Affiliation(s)
- Manuel Gámez-Chiachio
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - David Sarrió
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
- MD Anderson International Foundation, 28033 Madrid, Spain
| |
Collapse
|
19
|
Kankeu Fonkoua LA, Sirpilla O, Sakemura R, Siegler EL, Kenderian SS. CAR T cell therapy and the tumor microenvironment: Current challenges and opportunities. Mol Ther Oncolytics 2022; 25:69-77. [PMID: 35434273 PMCID: PMC8980704 DOI: 10.1016/j.omto.2022.03.009] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has demonstrated remarkable outcomes in individuals with hematological malignancies, but its success has been hindered by barriers intrinsic to the tumor microenvironment (TME), particularly for solid tumors, where it has yet to make its mark. In this article, we provide an updated review and future perspectives on features of the TME that represent barriers to CART cell therapy efficacy, including competition for metabolic fuels, physical barriers to infiltration, and immunosuppressive factors. We then discuss novel and promising strategies to overcome these obstacles that are in preclinical development or under clinical investigation.
Collapse
Affiliation(s)
- Lionel A. Kankeu Fonkoua
- T Cell Engineering Laboratory, Mayo Clinic, Rochester, MN, USA
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Olivia Sirpilla
- T Cell Engineering Laboratory, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Reona Sakemura
- T Cell Engineering Laboratory, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Elizabeth L. Siegler
- T Cell Engineering Laboratory, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Saad S. Kenderian
- T Cell Engineering Laboratory, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
20
|
Agostinetto E, Montemurro F, Puglisi F, Criscitiello C, Bianchini G, Del Mastro L, Introna M, Tondini C, Santoro A, Zambelli A. Immunotherapy for HER2-Positive Breast Cancer: Clinical Evidence and Future Perspectives. Cancers (Basel) 2022; 14:2136. [PMID: 35565264 PMCID: PMC9105460 DOI: 10.3390/cancers14092136] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common malignancy among women worldwide, and HER2-positive breast cancer accounts for approximately 15% of all breast cancer diagnoses. The advent of HER2-targeting therapies has dramatically improved the survival of these patients, significantly reducing their risk of recurrence and death. However, as a significant proportion of patients ultimately develop resistance to these therapies, it is extremely important to identify new treatments to further improve their clinical outcomes. Immunotherapy has revolutionized the treatment and history of several cancer types, and it has already been approved as a standard of care for patients with triple-negative breast cancer. Based on a strong preclinical rationale, immunotherapy in HER2-positive breast cancer represents an intriguing field that is currently under clinical investigation. There is a close interplay between HER2-targeting therapies (both approved and under investigation) and the immune system, and several new immunotherapeutic strategies, including immune checkpoint inhibitors, CAR-T cells and therapeutic vaccines, are being studied in this disease. In this narrative review, we discuss the clinical evidence and the future perspectives of immunotherapy for patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Elisa Agostinetto
- Academic Trials Promoting Team, Institut Jules Bordet, L’Université Libre de Bruxelles (U.L.B), 1070 Brussels, Belgium;
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Filippo Montemurro
- Direzione Breast Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Fabio Puglisi
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy;
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Carmen Criscitiello
- Division of Early Drug Development, European Institute of Oncology IRCCS, 20141 Milan, Italy;
- Department of Oncology and Hematology, University of Milan, 20122 Milan, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Lucia Del Mastro
- IRCCS Ospedale Policlinico San Martino, Clinica di Oncologia Medica, 16132 Genova, Italy;
- Dipartimento di Medicina Interna e Specialità Medica, Università di Genova, 16124 Genova, Italy
| | - Martino Introna
- UOS Centro di Terapia Cellulare “G. Lanzani”, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy;
| | - Carlo Tondini
- Medical Oncology Unit, ASST Papa Giovanni XXIII, Piazza OMS 1, 27100 Bergamo, Italy;
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Humanitas Cancer Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
21
|
Dubois VP, Sehl OC, Foster PJ, Ronald JA. Visualizing CAR-T cell Immunotherapy Using 3 Tesla Fluorine-19 MRI. Mol Imaging Biol 2022; 24:298-308. [PMID: 34786668 PMCID: PMC8983548 DOI: 10.1007/s11307-021-01672-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/02/2021] [Accepted: 10/20/2021] [Indexed: 01/19/2023]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T cell cancer immunotherapies have shown remarkable results in patients with hematological malignancies and represent the first approved genetically modified cellular therapies. However, not all blood cancer patients respond favorably, serious side effects have been reported, and the treatment of solid tumors has been a challenge. An imaging tool for visualizing the variety of CAR-T cell products in use and being explored could provide important patient-specific data on CAR-T cell location to inform on potential success or failure of treatment as well as off-target toxicities. Fluorine-19 (19F) magnetic resonance imaging (MRI) allows for the noninvasive detection of 19F perfluorocarbon (PFC) labeled cells. Our objective was to visualize PFC-labeled (PFC +) CAR-T cells in a mouse model of leukemia using clinical field strength (3 Tesla) 19F MRI and compare the cytotoxicity of PFC + versus unlabeled CAR-T cells. PROCEDURES NSG mice (n = 17) received subcutaneous injections of CD19 + human B cell leukemia cells (NALM6) expressing firefly luciferase in their left hind flank (1 × 106). Twenty-one days later, each mouse received an intratumoral injection of 10 × 106 PFC + CD19-targeted CAR-T cells (n = 6), unlabeled CD19-targeted CAR-T cells (n = 3), PFC + untransduced T cells (n = 5), or an equivalent volume of saline (n = 3). 19F MRI was performed on mice treated with PFC + CAR-T cells days 1, 3, and 7 post-treatment. Bioluminescence imaging (BLI) was performed on all mice days - 1, 5, 10, and 14 post-treatment to monitor tumor response. RESULTS PFC + CAR-T cells were successfully detected in tumors using 19F MRI on days 1, 3, and 7 post-injection. In vivo BLI data revealed that mice treated with PFC + or PFC - CAR-T cells had significantly lower tumor burden by day 14 compared to untreated mice and mice treated with PFC + untransduced T cells (p < 0.05). Importantly, mice treated with PFC + CAR-T cells showed equivalent cytotoxicity compared to mice receiving PFC - CAR-T cells. CONCLUSIONS Our studies demonstrate that clinical field strength 19F MRI can be used to visualize PFC + CAR-T cells for up to 7 days post-intratumoral injection. Importantly, PFC labeling did not significantly affect in vivo CAR-T cell cytotoxicity. These imaging tools may have broad applications for tracking emerging CAR-T cell therapies in preclinical models and may eventually be useful for the detection of CAR-T cells in patients where localized injection of CAR-T cells is being pursued.
Collapse
Affiliation(s)
- Veronica P Dubois
- Robarts Research Institute, London, ON, Canada
- The Department of Medical Biophysics, Western University, London, ON, Canada
| | - Olivia C Sehl
- Robarts Research Institute, London, ON, Canada
- The Department of Medical Biophysics, Western University, London, ON, Canada
| | - Paula J Foster
- Robarts Research Institute, London, ON, Canada
- The Department of Medical Biophysics, Western University, London, ON, Canada
| | - John A Ronald
- Robarts Research Institute, London, ON, Canada.
- The Department of Medical Biophysics, Western University, London, ON, Canada.
- Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
22
|
Preclinical pharmacology modeling of chimeric antigen receptor T therapies. Curr Opin Pharmacol 2021; 61:49-61. [PMID: 34619442 DOI: 10.1016/j.coph.2021.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have largely been successful in treating hematological malignancies in the clinic but have not been as effective in treating solid tumors, in part, owing to poor access and the immunosuppressive tumor microenvironment. In addition, CAR-T therapy can cause potentially life-threatening side effects, including cytokine release syndrome and neurotoxicity. Current preclinical testing of CAR-T therapy efficacy is typically performed in mouse tumor models, which often fails to predict toxicity. Recent developments in humanized models and transgenic mice as well as in vitro three-dimensional organoids in early development and nonhuman primate models are being adopted for CAR-T cell efficacy and toxicity assessment. However, because no single model perfectly recapitulates the human immune system and tumor microenvironment, careful model selection based on their respective pros and cons is crucial for adequate evaluation of different CAR-T treatments, so that their clinical development can be better supported.
Collapse
|
23
|
Aharon A, Horn G, Bar-Lev TH, Zagagi Yohay E, Waks T, Levin M, Deshet Unger N, Avivi I, Globerson Levin A. Extracellular Vesicles Derived from Chimeric Antigen Receptor-T Cells: A Potential Therapy for Cancer. Hum Gene Ther 2021; 32:1224-1241. [PMID: 34494460 DOI: 10.1089/hum.2021.192] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells are genetically engineered T cells, directed against a tumor-associated antigen. Extracellular vesicles (EVs) derived from CAR-T cells (CAR-T EVs) may preserve CAR-T activity and overcome one of the major obstacles responsible for CAR-T cell failure in patients with solid tumors. This study aimed to compare CAR-T EVs with their parental cells and explore their cell penetration and cytotoxic activity. Anti-HER-2 CARs were stimulated with specific target cells. EVs were isolated from the cell media and characterized for their content and functions. We found that CAR-T EVs contained a mixture of small and large EVs. Stimulated anti-HER-2+ CAR-T EVs expressed lower cytokine levels compared with their parental CAR-T cells (such as interferon gamma). Higher levels of granzyme B were found in CAR-T EVs (≥20 × ) compared with EVs from unstimulated cells (p < 0.001). Anti-HER-2+ CAR-T EVs bound and penetrated specifically into HER-2 expressing target cells. Similar cytotoxic effects measured by caspase-3/7 activity were found in CAR-T cells and their derived EVs. However, while the CAR-T cells induced massive apoptosis during the first 24 h, CAR-T EVs required 60 - 90 h. In summary, CAR-T EVs provide a novel potent immunotherapy approach that may be effective against solid tumors.
Collapse
Affiliation(s)
- Anat Aharon
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galit Horn
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tali Hana Bar-Lev
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Einav Zagagi Yohay
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tova Waks
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Immunology Department, The Weizmann Institute, Rehovot, Israel
| | - Maya Levin
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Naamit Deshet Unger
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Irit Avivi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anat Globerson Levin
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Dotan Center for Advanced Therapies, Tel-Aviv Sourasky Medical Center and Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
24
|
Diamant G, Simchony Goldman H, Gasri Plotnitsky L, Roitman M, Shiloach T, Globerson-Levin A, Eshhar Z, Haim O, Pencovich N, Grossman R, Ram Z, Volovitz I. T Cells Retain Pivotal Antitumoral Functions under Tumor-Treating Electric Fields. THE JOURNAL OF IMMUNOLOGY 2021; 207:709-719. [PMID: 34215656 DOI: 10.4049/jimmunol.2100100] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/28/2021] [Indexed: 11/19/2022]
Abstract
Tumor-treating fields (TTFields) are a localized, antitumoral therapy using alternating electric fields, which impair cell proliferation. Combining TTFields with tumor immunotherapy constitutes a rational approach; however, it is currently unknown whether TTFields' locoregional effects are compatible with T cell functionality. Healthy donor PBMCs and viably dissociated human glioblastoma samples were cultured under either standard or TTFields conditions. Select pivotal T cell functions were measured by multiparametric flow cytometry. Cytotoxicity was evaluated using a chimeric Ag receptor (CAR)-T-based assay. Glioblastoma patient samples were acquired before and after standard chemoradiation or standard chemoradiation + TTFields treatment and examined by immunohistochemistry and by RNA sequencing. TTFields reduced the viability of proliferating T cells, but had little or no effect on the viability of nonproliferating T cells. The functionality of T cells cultured under TTFields was retained: they exhibited similar IFN-γ secretion, cytotoxic degranulation, and PD1 upregulation as controls with similar polyfunctional patterns. Glioblastoma Ag-specific T cells exhibited unaltered viability and functionality under TTFields. CAR-T cells cultured under TTFields exhibited similar cytotoxicity as controls toward their CAR target. Transcriptomic analysis of patients' glioblastoma samples revealed a significant shift in the TTFields-treated versus the standard-treated samples, from a protumoral to an antitumoral immune signature. Immunohistochemistry of samples before and after TTFields treatment showed no reduction in T cell infiltration. T cells were found to retain key antitumoral functions under TTFields settings. Our data provide a mechanistic insight and a rationale for ongoing and future clinical trials that combine TTFields with immunotherapy.
Collapse
Affiliation(s)
- Gil Diamant
- The Cancer Immunotherapy Laboratory, Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University Tel-Aviv, Israel.,Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Hadar Simchony Goldman
- The Cancer Immunotherapy Laboratory, Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University Tel-Aviv, Israel
| | - Lital Gasri Plotnitsky
- The Cancer Immunotherapy Laboratory, Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University Tel-Aviv, Israel
| | - Marina Roitman
- The Cancer Immunotherapy Laboratory, Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University Tel-Aviv, Israel
| | - Tamar Shiloach
- Laboratory for Cancer Research and Immunotherapy, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Anat Globerson-Levin
- Laboratory for Cancer Research and Immunotherapy, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Zelig Eshhar
- Laboratory for Cancer Research and Immunotherapy, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Oz Haim
- Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Niv Pencovich
- Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Rachel Grossman
- Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Zvi Ram
- Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Ilan Volovitz
- The Cancer Immunotherapy Laboratory, Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University Tel-Aviv, Israel; .,Neurosurgery Department, Tel-Aviv Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| |
Collapse
|
25
|
Rousso-Noori L, Mastandrea I, Talmor S, Waks T, Globerson Levin A, Haugas M, Teesalu T, Alvarez-Vallina L, Eshhar Z, Friedmann-Morvinski D. P32-specific CAR T cells with dual antitumor and antiangiogenic therapeutic potential in gliomas. Nat Commun 2021; 12:3615. [PMID: 34127674 PMCID: PMC8203650 DOI: 10.1038/s41467-021-23817-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 05/12/2021] [Indexed: 12/29/2022] Open
Abstract
Glioblastoma is considered one of the most aggressive malignancies in adult and pediatric patients. Despite decades of research no curative treatment is available and it thus remains associated with a very dismal prognosis. Although recent pre-clinical and clinical studies have demonstrated the feasibility of chimeric antigen receptors (CAR) T cell immunotherapeutic approach in glioblastoma, tumor heterogeneity and antigen loss remain among one of the most important challenges to be addressed. In this study, we identify p32/gC1qR/HABP/C1qBP to be specifically expressed on the surface of glioma cells, making it a suitable tumor associated antigen for redirected CAR T cell therapy. We generate p32 CAR T cells and find them to recognize and specifically eliminate p32 expressing glioma cells and tumor derived endothelial cells in vitro and to control tumor growth in orthotopic syngeneic and xenograft mouse models. Thus, p32 CAR T cells may serve as a therapeutic option for glioblastoma patients. Chimeric antigen receptor (CAR) T cell therapy has been proposed as a promising approach for treating glioblastoma. Here the authors show that p32 is expressed in murine and human glioma and that p32-directed CAR-T cells promote anti-tumor responses in preclinical models by targeting glioma cells and tumor derived endothelial cells.
Collapse
Affiliation(s)
- Liat Rousso-Noori
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ignacio Mastandrea
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shauli Talmor
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tova Waks
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Maarja Haugas
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia.,Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Luis Alvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.,Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| | - Zelig Eshhar
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dinorah Friedmann-Morvinski
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
26
|
Fuentes-Antrás J, Guevara-Hoyer K, Baliu-Piqué M, García-Sáenz JÁ, Pérez-Segura P, Pandiella A, Ocaña A. Adoptive Cell Therapy in Breast Cancer: A Current Perspective of Next-Generation Medicine. Front Oncol 2020; 10:605633. [PMID: 33194771 PMCID: PMC7653090 DOI: 10.3389/fonc.2020.605633] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy has become a cornerstone in the treatment of cancer and changed the way clinicians and researchers approach tumor vulnerabilities. Durable responses are commonly observed with immune checkpoint inhibitors in highly immunogenic tumors, while the infusion of T cells genetically engineered to express chimeric antigen receptors (CARs) has shown impressive efficacy in certain types of blood cancer. Nevertheless, harnessing our own immunity has not proved successful for most breast cancer patients. In the era of genomic medicine, cellular immunotherapies may provide a more personalized and dynamic tool against tumors displaying heterogeneous mutational landscapes and antigenic pools. This approach encompasses multiple strategies including the adoptive transfer of tumor-infiltrating lymphocytes, dendritic cells, natural killer cells, and engineered immune components such as CAR constructs and engineered T cell receptors. Although far from permeating the clinical setting, technical advances have been overwhelming in recent years, with continuous improvement in traditional challenges such as toxicity, adoptive cell persistence, and intratumoral trafficking. Also, there is an avid search for neoantigens that can be targeted by these strategies, either alone or in combination. In this work, we aim to provide a clinically-oriented overview of preclinical and clinical data regarding the use of cellular immunotherapies in breast cancer.
Collapse
Affiliation(s)
- Jesús Fuentes-Antrás
- Breast Cancer Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain.,Experimental Therapeutics and Translational Oncology Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| | - Kissy Guevara-Hoyer
- Clinical Immunology Department, San Carlos University Hospital, Madrid, Spain
| | - Mariona Baliu-Piqué
- Experimental Therapeutics and Translational Oncology Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| | - José Ángel García-Sáenz
- Breast Cancer Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| | - Pedro Pérez-Segura
- Breast Cancer Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain.,Experimental Therapeutics and Translational Oncology Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| | - Atanasio Pandiella
- Institute of Molecular and Cellular Biology of Cancer and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC), Salamanca, Spain
| | - Alberto Ocaña
- Breast Cancer Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain.,Experimental Therapeutics and Translational Oncology Unit, Medical Oncology Department, San Carlos University Hospital, Madrid, Spain
| |
Collapse
|
27
|
Globerson Levin A, Rawet Slobodkin M, Waks T, Horn G, Ninio-Many L, Deshet Unger N, Ohayon Y, Suliman S, Cohen Y, Tartakovsky B, Naparstek E, Avivi I, Eshhar Z. Treatment of Multiple Myeloma Using Chimeric Antigen Receptor T Cells with Dual Specificity. Cancer Immunol Res 2020; 8:1485-1495. [PMID: 33008840 DOI: 10.1158/2326-6066.cir-20-0118] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/04/2020] [Accepted: 09/29/2020] [Indexed: 11/16/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has shown remarkable successes in fighting B-cell leukemias/lymphomas. Promising response rates are reported in patients treated with B-cell maturation antigen (BCMA) CAR T cells for multiple myeloma. However, responses appear to be nondurable, highlighting the need to expand the repertoire of multiple myeloma-specific targets for immunotherapy and to generate new CAR T cells. Here, we developed a "dual-CAR" targeting two multiple myeloma-associated antigens and explored its safety and efficacy. To reduce the "off-target" toxicity, we used the recognition of paired antigens that were coexpressed by the tumor to induce efficient CAR T-cell activation. The dual-CAR construct presented here was carefully designed to target the multiple myeloma-associated antigens, taking into consideration the distribution of both antigens on normal human tissues. Our results showed that the CD138/CD38-targeted dual CAR (dCAR138-38) elicited a potent anti-multiple myeloma response both in vitro and in vivo NSG mice transplanted with a multiple myeloma cell line and treated with dCAR138-38 showed median survival of 97 days compared with 31 days in the control group treated with mock-lymphocytes. The dCAR138-38 showed increased specificity toward cells expressing both targeted antigens compared with single-antigen-expressing cells and low activity toward primary cells from healthy tissues. Our findings indicated that the dCAR138-38 may provide a potent and safe alternative therapy for patients with multiple myeloma.
Collapse
Affiliation(s)
- Anat Globerson Levin
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel. .,Weizmann Institute of Science, Rehovot, Israel
| | | | - Tova Waks
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Weizmann Institute of Science, Rehovot, Israel
| | - Galit Horn
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel
| | | | | | - Yaara Ohayon
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel
| | | | - Yael Cohen
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Ella Naparstek
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel
| | - Irit Avivi
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zelig Eshhar
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Weizmann Institute of Science, Rehovot, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Szöőr Á, Tóth G, Zsebik B, Szabó V, Eshhar Z, Abken H, Vereb G. Trastuzumab derived HER2-specific CARs for the treatment of trastuzumab-resistant breast cancer: CAR T cells penetrate and eradicate tumors that are not accessible to antibodies. Cancer Lett 2020; 484:1-8. [DOI: 10.1016/j.canlet.2020.04.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/26/2020] [Accepted: 04/04/2020] [Indexed: 12/18/2022]
|
29
|
Chulpanova DS, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV. Mouse Tumor Models for Advanced Cancer Immunotherapy. Int J Mol Sci 2020; 21:E4118. [PMID: 32526987 PMCID: PMC7312663 DOI: 10.3390/ijms21114118] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022] Open
Abstract
Recent advances in the development of new methods of cancer immunotherapy require the production of complex cancer animal models that reliably reflect the complexity of the tumor and its microenvironment. Mice are good animals to create tumor models because they are low cost, have a short reproductive cycle, exhibit high tumor growth rates, and can be easily genetically modified. However, the obvious problem of these models is the high failure rate observed in human clinical trials after promising results obtained in mouse models. In order to increase the reliability of the results obtained in mice, the tumor model should reflect the heterogeneity of the tumor, contain components of the tumor microenvironment, in particular immune cells, to which the action of immunotherapeutic drugs are directed. This review discusses the current immunocompetent and immunocompromised mouse models of human tumors that are used to evaluate the effectiveness of immunotherapeutic agents, in particular chimeric antigen receptor (CAR) T-cells and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Daria S. Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Catrin S. Rutland
- Faculty of Medicine and Health Sciences, University of Medicine, Nottingham NG7 2HA, UK;
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (D.S.C.); (K.V.K.); (A.A.R.)
| |
Collapse
|
30
|
Tarantino P, Morganti S, Curigliano G. Biologic therapy for advanced breast cancer: recent advances and future directions. Expert Opin Biol Ther 2020; 20:1009-1024. [PMID: 32255704 DOI: 10.1080/14712598.2020.1752176] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Advanced breast cancer (ABC) is a leading cause of mortality, morbidity, and disability in women worldwide. For decades, treatment of ABC has relied on chemotherapy and endocrine treatments (ET), until HER2 was recognized as a 'druggable' target in the 1990s. Thereafter, various anti-HER2 drugs have been approved for the HER2-positive subtype, but only in the last few years, biologic agents targeting different pathways have entered the therapeutic arsenal of luminal and triple-negative cancers. AREAS COVERED The purpose of the present review is to recapitulate the most promising novel biologic agents being developed for the treatment of ABC. New drugs for all breast cancer subtypes are discussed, as well as some potential future directions in ABC treatment. EXPERT OPINION Several biologic drugs have been recently approved, revolutionizing ABC treatment algorithms: key examples are CDK4/6-inhibitors and the PI3K-inhibitor alpelisib for endocrine-positive ABC; atezolizumab for triple-negative cancers; two PARP-inhibitors for HER2-negative germinal BRCA-mutated cancers. Additionally, multiple drugs are demonstrating activity in late-phase clinical trials for all subtypes. While some of these represent pharmacological evolutions of previously approved drugs, some others might pave the way for new paradigms in ABC, challenging both its classification and current treatment algorithms.
Collapse
Affiliation(s)
- Paolo Tarantino
- European Institute of Oncology, IRCCS, Division of Early Drug Development for Innovative Therapies , Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan, Italy
| | - Stefania Morganti
- European Institute of Oncology, IRCCS, Division of Early Drug Development for Innovative Therapies , Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan, Italy
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Division of Early Drug Development for Innovative Therapies , Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan , Milan, Italy
| |
Collapse
|
31
|
Less is more: reducing the number of administered chimeric antigen receptor T cells in a mouse model using a mathematically guided approach. Cancer Immunol Immunother 2020; 69:1165-1175. [DOI: 10.1007/s00262-020-02516-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/12/2020] [Indexed: 12/13/2022]
|
32
|
Tóth G, Szöllősi J, Abken H, Vereb G, Szöőr Á. A Small Number of HER2 Redirected CAR T Cells Significantly Improves Immune Response of Adoptively Transferred Mouse Lymphocytes against Human Breast Cancer Xenografts. Int J Mol Sci 2020; 21:ijms21031039. [PMID: 32033208 PMCID: PMC7038081 DOI: 10.3390/ijms21031039] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 12/17/2022] Open
Abstract
HER2 positive JIMT-1 breast tumors are resistant to trastuzumab treatment in vitro and develop resistance to trastuzumab in vivo in SCID mice. We explored whether these resistant tumors could still be eliminated by T cells redirected by a second-generation chimeric antigen receptor (CAR) containing a CD28 costimulatory domain and targeting HER2 with a trastuzumab-derived scFv. In vitro, T cells engineered with this HER2 specific CAR recognized HER2 positive target cells as judged by cytokine production and cytolytic activity. In vivo, the administration of trastuzumab twice weekly had no effect on the growth of JIMT-1 xenografts in SCID mice. At the same time, a single dose of 2.5 million T cells from congenic mice exhibited a moderate xenoimmune response and even stable disease in some cases. In contrast, when the same dose contained 7% (175,000) CAR T cells, complete remission was achieved in 57 days. Even a reduced dose of 250,000 T cells, including only 17,500 CAR T cells, yielded complete remission, although it needed nearly twice the time. We conclude that even a small number of CAR T lymphocytes can evoke a robust anti-tumor response against an antibody resistant xenograft by focusing the activity of xenogenic T cells. This observation may have significance for optimizing the dose of CAR T cells in the therapy of solid tumors.
Collapse
Affiliation(s)
- Gábor Tóth
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, 4032 Debrecen, Hungary; (G.T.); (J.S.)
| | - János Szöllősi
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, 4032 Debrecen, Hungary; (G.T.); (J.S.)
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Hinrich Abken
- Regensburg Center for Interventional Immunology, Dept. Genetic Immunotherapy, and University Hospital Regensburg, D-93053 Regensburg, Germany;
| | - György Vereb
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, 4032 Debrecen, Hungary; (G.T.); (J.S.)
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (G.V.); (A.S.); Tel.: +36-52-258-603 (G.V. & A.S.)
| | - Árpád Szöőr
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, 4032 Debrecen, Hungary; (G.T.); (J.S.)
- Correspondence: (G.V.); (A.S.); Tel.: +36-52-258-603 (G.V. & A.S.)
| |
Collapse
|
33
|
Marei HE, Althani A, Caceci T, Arriga R, Sconocchia T, Ottaviani A, Lanzilli G, Roselli M, Caratelli S, Cenciarelli C, Sconocchia G. Recent perspective on CAR and Fcγ-CR T cell immunotherapy for cancers: Preclinical evidence versus clinical outcomes. Biochem Pharmacol 2019; 166:335-346. [PMID: 31176617 DOI: 10.1016/j.bcp.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
The chimeric antigen receptor T cell (CAR-T cell) immunotherapy currently represents a hot research trend and it is expected to revolutionize the field of cancer therapy. Promising outcomes have been achieved using CAR-T cell therapy for haematological malignancies. Despite encouraging results, several challenges still pose eminent hurdles before being fully recognized. Directing CAR-T cells to target a single tumour associated antigen (TAA) as the case in haematological malignancies might be much simpler than targeting the extensive inhibitory microenvironments associated with solid tumours. This review focuses on the basic principles involved in development of CAR-T cells, emphasizing the differences between humoral IgG, T-cell receptors, CAR and Fcγ-CR constructs. It also highlights the complex inhibitory network that is usually associated with solid tumours, and tackles recent advances in the clinical studies that have provided great hope for the future use of CAR-T cell immunotherapy. While current Fcγ-CR T cell immunotherapy is in pre-clinical stage, is expected to provide a sound therapeutic approach to add to existing classical chemo- and radio-therapeutic modalities.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt.
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Thomas Caceci
- Biomedical Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Roberto Arriga
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | | | | | - Mario Roselli
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Sara Caratelli
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | | | |
Collapse
|
34
|
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: 48] [Impact Index Per Article: 8.0] [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
|
35
|
Humanized Mice for the Study of Immuno-Oncology. Trends Immunol 2018; 39:748-763. [DOI: 10.1016/j.it.2018.07.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 01/28/2023]
|
36
|
Siegler EL, Wang P. Preclinical Models in Chimeric Antigen Receptor-Engineered T-Cell Therapy. Hum Gene Ther 2018; 29:534-546. [PMID: 29390873 DOI: 10.1089/hum.2017.243] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy has enormous potential in inducing long-term remission in cancer patients, and chimeric antigen receptor (CAR)-engineered T cells have been largely successful in treating hematological malignancies in the clinic. CAR-T therapy has not been as effective in treating solid tumors, in part due to the immunosuppressive tumor microenvironment. Additionally, CAR-T therapy can cause dangerous side effects, including off-tumor toxicity, cytokine release syndrome, and neurotoxicity. Animal models of CAR-T therapy often fail to predict such adverse events and frequently overestimate the efficacy of the treatment. Nearly all preclinical CAR-T studies have been performed in mice, including syngeneic, xenograft, transgenic, and humanized mouse models. Recently, a few studies have used primate models to mimic clinical side effects better. To date, no single model perfectly recapitulates the human immune system and tumor microenvironment, and some models have revealed CAR-T limitations that were contradicted or missed entirely in other models. Careful model selection based on the primary goals of the study is a crucial step in evaluating CAR-T treatment. Advancements are being made in preclinical models, with the ultimate objective of providing safer, more effective CAR-T therapy to patients.
Collapse
Affiliation(s)
- Elizabeth Louise Siegler
- 1 Department of Biomedical Engineering, University of Southern California , Los Angeles, California
| | - Pin Wang
- 1 Department of Biomedical Engineering, University of Southern California , Los Angeles, California.,2 Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , Los Angeles, California.,3 Mork Family Department of Chemical Engineering and Materials Science, University of Southern California , Los Angeles, California
| |
Collapse
|
37
|
Han Y, Liu C, Li G, Li J, Lv X, Shi H, Liu J, Liu S, Yan P, Wang S, Sun Y, Sun M. Antitumor effects and persistence of a novel HER2 CAR T cells directed to gastric cancer in preclinical models. Am J Cancer Res 2018; 8:106-119. [PMID: 29416924 PMCID: PMC5794725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 11/29/2017] [Indexed: 06/08/2023] Open
Abstract
New immunotherapeutic approaches are urgently needed for gastric cancer due to its poor survival and unsatisfactory treatment. Here we applied the humanized chA21 scfv based chimeric antigen receptor (CAR) modified T cells approach to the HER2 overexpressing gastric cancer treatment. The chA21-4-1BBz CAR T cells specifically exerted Th1 skewed cytokine response and efficient cytolysis of HER2 overexpressing human gastric cancer cells in vitro. Both the cytokine production and cytotoxicity levels were correlated with the level of HER2 surface expression by tumor cells. In established subcutaneous xenograft and peritoneal metastasis models, chA21-4-1BBz CAR T cells dramatically facilitated regression of HER2 overexpressing tumor and prolonged survival of tumor-bearing mice, whereas spared the progression of HER2 low-expressing tumor. Additionally, the capability of these CAR T cells to persist in circulation, as well as specifically home to, and accumulate in tumor sites were identified. Taken together, these results provide the basis for the future clinical investigation of the humanized chA21 scFv based, 4-1BB costimulated CAR T cells for the treatment of gastric cancer, and other HER2-expressing solid tumors.
Collapse
Affiliation(s)
- Yali Han
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Chuanyong Liu
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Guanhua Li
- Department of Respiration, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Juan Li
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Xingyan Lv
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Huan Shi
- Department of Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical SciencesJinan, Shandong, P. R. China
| | - Jie Liu
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Shuai Liu
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Peng Yan
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Shuyun Wang
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| | - Meili Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong UniversityJinan 250013, Shandong, P. R. China
| |
Collapse
|
38
|
Yeku O, Li X, Brentjens RJ. Adoptive T-Cell Therapy for Solid Tumors. AMERICAN SOCIETY OF CLINICAL ONCOLOGY EDUCATIONAL BOOK. AMERICAN SOCIETY OF CLINICAL ONCOLOGY. ANNUAL MEETING 2017. [PMID: 28561728 DOI: 10.14694/edbk_180328] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and various costimulatory molecules. Upon administration, these modified T cells traffic to, and recognize, cancer cells in an HLA-independent manner. CAR T-cell therapy has shown remarkable success in the treatment of CD-19-expressing B-cell acute lymphocytic leukemia. However, clinical gains to the same magnitude have not been reported in solid tumors. Several known obstacles to CAR T-cell therapy for solid tumors include target antigen identification, effective trafficking to the tumor, robust activation, proliferation, and in vivo cytotoxicity. Beyond these T-cell intrinsic properties, a complex and dynamic immunosuppressive tumor microenvironment in solid tumors hinders T-cell efficacy. Notable advancements in CAR design to include multiple costimulatory molecules, ligands, and soluble cytokines have shown promise in preclinical models, and some of these are currently in early-phase clinical trials. In this review, we discuss selected solid tumor malignancies and relevant preclinical data and highlight clinical trial results that are available. Furthermore, we outline some obstacles to CAR T-cell therapy for each tumor and propose strategies to overcome some of these limitations.
Collapse
Affiliation(s)
- Oladapo Yeku
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xinghuo Li
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
39
|
Yeku O, Li X, Brentjens RJ. Adoptive T-Cell Therapy for Solid Tumors. Am Soc Clin Oncol Educ Book 2017; 37:193-204. [PMID: 28561728 DOI: 10.1200/edbk_180328] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and various costimulatory molecules. Upon administration, these modified T cells traffic to, and recognize, cancer cells in an HLA-independent manner. CAR T-cell therapy has shown remarkable success in the treatment of CD-19-expressing B-cell acute lymphocytic leukemia. However, clinical gains to the same magnitude have not been reported in solid tumors. Several known obstacles to CAR T-cell therapy for solid tumors include target antigen identification, effective trafficking to the tumor, robust activation, proliferation, and in vivo cytotoxicity. Beyond these T-cell intrinsic properties, a complex and dynamic immunosuppressive tumor microenvironment in solid tumors hinders T-cell efficacy. Notable advancements in CAR design to include multiple costimulatory molecules, ligands, and soluble cytokines have shown promise in preclinical models, and some of these are currently in early-phase clinical trials. In this review, we discuss selected solid tumor malignancies and relevant preclinical data and highlight clinical trial results that are available. Furthermore, we outline some obstacles to CAR T-cell therapy for each tumor and propose strategies to overcome some of these limitations.
Collapse
Affiliation(s)
- Oladapo Yeku
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xinghuo Li
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
40
|
Sridhar P, Petrocca F. Regional Delivery of Chimeric Antigen Receptor (CAR) T-Cells for Cancer Therapy. Cancers (Basel) 2017; 9:E92. [PMID: 28718815 PMCID: PMC5532628 DOI: 10.3390/cancers9070092] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/08/2017] [Accepted: 07/11/2017] [Indexed: 11/16/2022] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cells are T-cells with recombinant receptors targeted to tumor antigens. CAR-T cell therapy has emerged as a mode of immunotherapy and is now being extensively explored in hematologic cancer. In contrast, CAR-T cell use in solid tumors has been hampered by multiple obstacles. Several approaches have been taken to circumvent these obstacles, including the regional delivery of CAR-T cells. Regional CAR-T cell delivery can theoretically compensate for poor T-cell trafficking and tumor antigen specificity while avoiding systemic toxicity associated with intravenous delivery. We reviewed completed clinical trials for the treatment of glioblastoma and metastatic colorectal cancer and examined the data in these studies for safety, efficacy, and potential advantages that regional delivery may confer over systemic delivery. Our appraisal of the available literature revealed that regional delivery of CAR-T cells in both glioblastoma and hepatic colorectal metastases was generally well tolerated and efficacious in select instances. We propose that the regional delivery of CAR-T cells is an area of potential growth in the solid tumor immunotherapy, and look towards future clinical trials in head and neck cancer, mesothelioma, and peritoneal carcinomatosis as the use of this technique expands.
Collapse
Affiliation(s)
- Praveen Sridhar
- Department of Surgery, Boston University, Boston, MA 02118, USA.
| | - Fabio Petrocca
- Department of Surgery, Boston University, Boston, MA 02118, USA.
| |
Collapse
|
41
|
Gross G, Eshhar Z. Therapeutic Potential of T Cell Chimeric Antigen Receptors (CARs) in Cancer Treatment: Counteracting Off-Tumor Toxicities for Safe CAR T Cell Therapy. Annu Rev Pharmacol Toxicol 2016; 56:59-83. [PMID: 26738472 DOI: 10.1146/annurev-pharmtox-010814-124844] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A chimeric antigen receptor (CAR) is a recombinant fusion protein combining an antibody-derived targeting fragment with signaling domains capable of activating T cells. Recent early-phase clinical trials have demonstrated the remarkable ability of CAR-modified T cells to eliminate B cell malignancies. This review describes the choice of target antigens and CAR manipulations to maximize antitumor specificity. Benefits and current limitations of CAR-modified T cells are discussed, with a special focus on the distribution of tumor antigens on normal tissues and the risk of on-target, off-tumor toxicities in the clinical setting. We present current methodologies for pre-evaluating these risks and review the strategies for counteracting potential off-tumor effects. Successful implementation of these approaches will improve the safety and efficacy of CAR T cell therapy and extend the range of cancer patients who may be treated.
Collapse
Affiliation(s)
- Gideon Gross
- Laboratory of Immunology, MIGAL, Galilee Research Institute, Kiryat Shmona 11016, Israel; .,Department of Biotechnology, Tel-Hai College, Upper Galilee 12210, Israel.,Center of Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
| | - Zelig Eshhar
- Center of Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel.,Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
42
|
Magee MS, Kraft CL, Abraham TS, Baybutt TR, Marszalowicz GP, Li P, Waldman SA, Snook AE. GUCY2C-directed CAR-T cells oppose colorectal cancer metastases without autoimmunity. Oncoimmunology 2016; 5:e1227897. [PMID: 27853651 PMCID: PMC5087292 DOI: 10.1080/2162402x.2016.1227897] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
Adoptive T-cell therapy (ACT) is an emerging paradigm in which T cells are genetically modified to target cancer-associated antigens and eradicate tumors. However, challenges treating epithelial cancers with ACT reflect antigen targets that are not tumor-specific, permitting immune damage to normal tissues, and preclinical testing in artificial xenogeneic models, preventing prediction of toxicities in patients. In that context, mucosa-restricted antigens expressed by cancers exploit anatomical compartmentalization which shields mucosae from systemic antitumor immunity. This shielding may be amplified with ACT platforms employing antibody-based chimeric antigen receptors (CARs), which mediate MHC-independent recog-nition of antigens. GUCY2C is a cancer mucosa antigen expressed on the luminal surfaces of the intestinal mucosa in mice and humans, and universally overexpressed by colorectal tumors, suggesting its unique utility as an ACT target. T cells expressing CARs directed by a GUCY2C-specific antibody fragment recognized GUCY2C, quantified by expression of activation markers and cytokines. Further, GUCY2C CAR-T cells lysed GUCY2C-expressing, but not GUCY2C-deficient, mouse colorectal cancer cells. Moreover, GUCY2C CAR-T cells reduced tumor number and morbidity and improved survival in mice harboring GUCY2C-expressing colorectal cancer metastases. GUCY2C-directed T cell efficacy reflected CAR affinity and surface expression and was achieved without immune-mediated damage to normal tissues in syngeneic mice. These observations highlight the potential for therapeutic translation of GUCY2C-directed CAR-T cells to treat metastatic tumors, without collateral autoimmunity, in patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Michael S Magee
- Bluebird Bio, Seattle, Cambridge, MA, USA; Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - Crystal L Kraft
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| | - Tara S Abraham
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| | - Trevor R Baybutt
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| | - Glen P Marszalowicz
- School of Biomedical Engineering, Science & Health Systems, Drexel University , Philadelphia, PA, USA
| | - Peng Li
- Department of Pathology, Stanford University School of Medicine , Stanford, CA, USA
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University , Philadelphia, PA, USA
| |
Collapse
|
43
|
Strain-dependent Lethal Toxicity in NKG2D Ligand-targeted CAR T-cell Therapy. Mol Ther 2016; 23:1559-61. [PMID: 26442803 DOI: 10.1038/mt.2015.162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
44
|
Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells. Leukemia 2015; 30:701-7. [PMID: 26526988 DOI: 10.1038/leu.2015.311] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/26/2015] [Indexed: 12/21/2022]
Abstract
Peripheral T-cell lymphomas (PTCLs) are aggressive lymphomas with no effective upfront standard treatment and ineffective options in relapsed disease, resulting in poorer clinical outcomes as compared with B-cell lymphomas. The adoptive transfer of T cells engineered to express chimeric antigen receptors (CARs) is a promising new approach for treatment of hematological malignancies. However, preclinical reports of targeting T-cell lymphoma with CARs are almost non-existent. Here we have designed a CAR, CD4CAR, which redirects the antigen specificity of CD8+ cytotoxic T cells to CD4-expressing cells. CD4CAR T cells derived from human peripheral blood mononuclear cells and cord blood effectively redirected T-cell specificity against CD4+ cells in vitro. CD4CAR T cells efficiently eliminated a CD4+ leukemic cell line and primary CD4+ PTCL patient samples in co-culture assays. Notably, CD4CAR T cells maintained a central memory stem cell-like phenotype (CD8+CD45RO+CD62L+) under standard culture conditions. Furthermore, in aggressive orthotropic T-cell lymphoma models, CD4CAR T cells efficiently suppressed the growth of lymphoma cells while also significantly prolonging mouse survival. Combined, these studies demonstrate that CD4CAR-expressing CD8+ T cells are efficacious in ablating malignant CD4+ populations, with potential use as a bridge to transplant or stand-alone therapy for the treatment of PTCLs.
Collapse
|
45
|
Nahas GR, Walker ND, Bryan M, Rameshwar P. A Perspective of Immunotherapy for Breast Cancer: Lessons Learned and Forward Directions for All Cancers. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:35-43. [PMID: 26568682 PMCID: PMC4631157 DOI: 10.4137/bcbcr.s29425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/09/2015] [Accepted: 07/12/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy for cancer has been a focus 50 years ago. At the time, this treatment was developed prior to cloning of the cytokines, no knowledge of regulatory T-cells, and very little information that mesenchymal stem cells (MSCs) (originally colony forming unit-fibroblasts [CFU-F]) could be licensed by the inflammatory microenvironment to suppress an immune response. Given the information available at that time, mononuclear cells from the peripheral blood were activated ex vivo and then replaced in the patients with tumor. The intent was to harness these activated immune cells to target the cancer cells. These studies did not lead to long-term responses because the activated cells when reinfused into the patients were an advantage to the resident MSCs, which can home the tumor and then become suppressive in the presence of the immune cells. The immune suppression caused by MSCs would also expand regulatory T-cells, resulting instead in tumor protection. As time progressed, these different fields converged into a new approach to use immunotherapy for cancer. This article discusses these approaches and also reviews chimeric antigen receptor in the context of future treatments for solid tumors, including breast cancer.
Collapse
Affiliation(s)
| | - Nykia D Walker
- Rutgers New Jersey Medical School, Newark, NJ, USA. ; Rutgers Graduate School of Biomedical Sciences, Newark, NJ, USA
| | | | - Pranela Rameshwar
- Rutgers New Jersey Medical School, Newark, NJ, USA. ; Rutgers Graduate School of Biomedical Sciences, Newark, NJ, USA
| |
Collapse
|
46
|
Eshhar Z. From the mouse cage to human therapy: a personal perspective of the emergence of T-bodies/chimeric antigen receptor T cells. Hum Gene Ther 2015; 25:773-8. [PMID: 25244568 DOI: 10.1089/hum.2014.2532] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zelig Eshhar
- Department of Immunology, The Weizmann Institute of Science , Rehovot, Israel 76100
| |
Collapse
|