101
|
Kirtane K, Elmariah H, Chung CH, Abate-Daga D. Adoptive cellular therapy in solid tumor malignancies: review of the literature and challenges ahead. J Immunother Cancer 2021; 9:jitc-2021-002723. [PMID: 34301811 PMCID: PMC8311333 DOI: 10.1136/jitc-2021-002723] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 01/01/2023] Open
Abstract
While immune checkpoint inhibitors (ICIs) have ushered in major changes in standards of care for many solid tumor malignancies, primary and acquired resistance is common. Insufficient antitumor T cells, inadequate function of these cells, and impaired formation of memory T cells all contribute to resistance mechanisms to ICI. Adoptive cellular therapy (ACT) is a form of immunotherapy that is rapidly growing in clinical investigation and has the potential to overcome these limitations by its ability to augment the number, specificity, and reactivity of T cells against tumor tissue. ACT has revolutionized the treatment of hematologic malignancies, though the use of ACT in solid tumor malignancies is still in its early stages. There are currently three major modalities of ACT: tumor-infiltrating lymphocytes (TILs), genetically engineered T-cell receptors (TCRs), and chimeric antigen receptor (CAR) T cells. TIL therapy involves expansion of a heterogeneous population of endogenous T cells found in a harvested tumor, while TCRs and CAR T cells involve expansion of a genetically engineered T-cell directed toward specific antigen targets. In this review, we explore the potential of ACT as a treatment modality against solid tumors, discuss their advantages and limitations against solid tumor malignancies, discuss the promising therapies under active investigation, and examine future directions for this rapidly growing field.
Collapse
Affiliation(s)
- Kedar Kirtane
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Hany Elmariah
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Daniel Abate-Daga
- Departments of Immunology, Cutaneous Oncology, and Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
102
|
Gharghani MS, Simonian M, Bakhtiari F, Ghaffari MH, Fazli G, Bayat AA, Negahdari B. Chimeric antigen receptor T-cell therapy for breast cancer. Future Oncol 2021; 17:2961-2979. [PMID: 34156280 DOI: 10.2217/fon-2020-1013] [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: 11/21/2022] Open
Abstract
One of the main reasons that researchers pay enormous attention to immunotherapy is that, despite significant advances in conventional therapy approaches, breast cancer remains the leading cause of death from malignant tumors among women. Genetically modifying T cells with chimeric antigen receptors (CAR) is one of the novel methods that has exhibited encouraging activity with relative safety, further urging investigators to develop several CAR T cells to target overexpressed antigens in breast tumors. This article is aimed not only to present such CAR T cells and discuss their remarkable results but also indicates their shortcomings with the hope of achieving possible strategies for improving therapeutic response.
Collapse
Affiliation(s)
- Mighmig Simonian Gharghani
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, 8415683111, Iran
| | - Miganoosh Simonian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| | - Faezeh Bakhtiari
- Department of Laboratory Sciences, Faculty of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, 71348-14336, Iran
| | - Mozhan Haji Ghaffari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| | - Ghazaleh Fazli
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ali Ahmad Bayat
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 14177-55469, Iran
| |
Collapse
|
103
|
Tahmasebi S, Elahi R, Khosh E, Esmaeilzadeh A. Programmable and multi-targeted CARs: a new breakthrough in cancer CAR-T cell therapy. Clin Transl Oncol 2021; 23:1003-1019. [PMID: 32997278 DOI: 10.1007/s12094-020-02490-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022]
Abstract
CAR-T cell therapy, as a novel immunotherapy approach, has indicated successful results in the treatment of hematological malignancies; however, distinct results have been achieved regarding solid tumors. Tumor immunosuppressive microenvironment has been identified as the most critical barrier in CAR-T cell therapy of solid tumors. Developing novel strategies to augment the safety and efficacy of CAR-T cells could be useful to overcome the solid tumor hurdles. Similar to other cancer treatments, CAR-T cell therapy can cause some side effects, which can disturb the healthy tissues. In the current review, we will discuss the practical breakthroughs in CAR-T cell therapy using the multi-targeted and programmable CARs instead of conventional types. These superior types of CAR-T cells have been developed to increase the function and safety of T cells in a controllable manner, which would diminish the incidence of relevant side effects. Moreover, we will describe the capability of these powerful CARs in targeting multiple tumor antigens, redirecting the CAR-T cells to specific target cells, incrementing the safety of CARs, and other advantages that lead to promising outcomes in cancer CAR-T cell therapy.
Collapse
Affiliation(s)
- S Tahmasebi
- Department of Immunology, Health Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - R Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - E Khosh
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - A Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Science, Zanjan, Iran.
- Cancer Gene Therapy Research Center, Zanjan University of Medical Science, Zanjan, Iran.
- Immunotherapy Research and Technology Group, Zanjan University of Medical Science, Zanjan, Iran.
| |
Collapse
|
104
|
Hull CM, Maher J. Approaches for refining and furthering the development of CAR-based T cell therapies for solid malignancies. Expert Opin Drug Discov 2021; 16:1105-1117. [PMID: 34038292 DOI: 10.1080/17460441.2021.1929920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Chimeric antigen receptor-engineered T-cells typically use the binding domains of antibodies to target cytotoxicity toward tumors. This approach has produced great efficacy against selected hematological cancers, but benefit in solid tumors has been limited. Characteristically, the microenvironment in solid tumors restricts CAR T cell function, thereby limiting success. Enhancing efficacy will depend on novel target discovery to refine specificity and reduce toxicity. Additionally, overcoming immunosuppressive mechanisms may be achieved by altering the structure of the CAR itself, together with ancillary gene expression or additional therapeutic interventions.Areas covered: Herein, the authors discuss approaches for refining and further developing CAR T cell therapies specifically for use with solid malignancies. The authors survey the existing literature and provide perspectives for the future.Expert opinion: Pronounced efficacy in solid tumors will likely require combination therapies, targeting both the tumor itself and associated immunosuppressive mechanisms. Future exploration of CAR T cell therapies for solid tumors is likely to incorporate next-generation designs that couple more precise targeting of cancer-associated targets with enhanced potency and resistance to exhaustion.
Collapse
Affiliation(s)
| | - John Maher
- King's College London, Division of Cancer Studies, Guy's Hospital, London, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, UK.,Department of Immunology, Eastbourne Hospital, East Sussex, UK
| |
Collapse
|
105
|
Glover M, Avraamides S, Maher J. How Can We Engineer CAR T Cells to Overcome Resistance? Biologics 2021; 15:175-198. [PMID: 34040345 PMCID: PMC8141613 DOI: 10.2147/btt.s252568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved unrivalled success in the treatment of B cell and plasma cell malignancies, with five CAR T cell products now approved by the US Food and Drug Administration (FDA). However, CAR T cell therapies for solid tumours have not been nearly as successful, owing to several additional challenges. Here, we discuss mechanisms of tumour resistance in CAR T cell therapy and the emerging strategies that are under development to engineer CAR T cells to overcome resistance.
Collapse
Affiliation(s)
- Maya Glover
- Leucid Bio Ltd., Guy's Hospital, London, SE1 9RT, UK
| | - Stephanie Avraamides
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, SE1 9RT, UK
| | - John Maher
- Leucid Bio Ltd., Guy's Hospital, London, SE1 9RT, UK.,King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, SE1 9RT, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK.,Department of Immunology, Eastbourne Hospital, Eastbourne, East Sussex, BN21 2UD, UK
| |
Collapse
|
106
|
Acute Myeloid Leukemia: Is It T Time? Cancers (Basel) 2021; 13:cancers13102385. [PMID: 34069204 PMCID: PMC8156992 DOI: 10.3390/cancers13102385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease driven by impaired differentiation of hematopoietic primitive cells toward myeloid lineages (monocytes, granulocytes, red blood cells, platelets), leading to expansion and accumulation of "stem" and/or "progenitor"-like or differentiated leukemic cells in the bone marrow and blood. AML progression alters the bone marrow microenvironment and inhibits hematopoiesis' proper functioning, causing sustained cytopenia and immunodeficiency. This review describes how the AML microenvironment influences lymphoid lineages, particularly T lymphocytes that originate from the thymus and orchestrate adaptive immune response. We focus on the elderly population, which is mainly affected by this pathology. We discuss how a permissive AML microenvironment can alter and even worsen the thymic function, T cells' peripheral homeostasis, phenotype, and functions. Based on the recent findings on the mechanisms supporting that AML induces quantitative and qualitative changes in T cells, we suggest and summarize current immunotherapeutic strategies and challenges to overcome these anomalies to improve the anti-leukemic immune response and the clinical outcome of patients.
Collapse
|
107
|
Liu Y, He Y. A narrative review of chimeric antigen receptor-T (CAR-T) cell therapy for lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:808. [PMID: 34268421 PMCID: PMC8246176 DOI: 10.21037/atm-20-5419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 03/14/2021] [Indexed: 01/22/2023]
Abstract
Lung cancer represents one of the most common and deadliest cancers in the world. Chimeric antigen receptor-T cell (CAR-T) therapy which can recognize antigens in a major histocompatibility complex (MHC)-independent manner provides a new approach for tumor treatment. However, lung cancer, as a solid tumor, faces several formidable barriers to adoptive cell transfer, which includes inhibition of T-cell localization and suppression of T-cell function. Therefore, lung cancer fails to respond significantly to infusions of CAR-T cells in most trials until now. PubMed was researched using the terms “CAR-T” and “lung cancer” only in English from 2000 through June 2020. We also included results presented in international conferences, such as the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO). Besides, we found new progress in CAR-T therapy for solid tumors as a supplement. To enhance the efficacy and conquer the limitations, we collected some applications in lung cancer. In recent years, there have been some improvements in selecting the proper target and reducing toxicity. CAR-T technology provides an excellent way for tumor treatment, which does not depend on MHC molecules and provides a new method for the utilization of tumor targets. Targeting different antigens and overcoming the solid barrier, there are some improvements in responding significantly and reducing toxicity. CAR-T technology will play a decisive role in the treatment of lung cancer.
Collapse
Affiliation(s)
| | - Yayi He
- Tongji University, Shanghai, China.,Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
108
|
Abstract
Breast cancer, as a heterogeneous disease, includes a wide range of pathological and clinical behaviors. Current treatment protocols, including radiotherapy, chemotherapy, and hormone replacement therapy, are mainly associated with poor response and high rate of recurrence. Therefore, more efforts are needed to develop alternative therapies for this type of cancer. Immunotherapy, as a novel strategy in cancer treatment, has a potential in treating breast cancer patients. Although breast cancer has long been considered problematic to treat with immunotherapy, as it is immunologically "cold," numerous newer preclinical and clinical reports now recommend that immunotherapy has the capability to treat breast cancer patients. In this review, we highlight the different immunotherapy strategies in breast cancer treatment.
Collapse
|
109
|
Holzinger A, Abken H. [Chimeric antigen receptors (CARs): universal tools in adoptive cell therapy]. Internist (Berl) 2021; 62:583-588. [PMID: 33928413 DOI: 10.1007/s00108-021-01040-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND The observation that tumor-infiltrating lymphocytes (TIL) after ex vivo amplification can control tumors in the long term led to the concept of redirecting patients' cytolytic T‑cells by a receptor with defined specificity against the tumor. OBJECTIVES Development of a recombinant receptor-signal molecule (chimeric antigen receptor, CAR) to increase selectivity and enhance anti-tumor immunity. METHODS Description of a prototype CAR, overview of the modular composition and further development of CAR technology for use in adoptive immune cell therapy. RESULTS Intensive research over the last two decades has shown how CAR-mediated T‑cell activation is influenced by factors such as binding affinity, the epitope of the target antigen, its expression density and accessibility on the tumor cells, as well as by the signaling domains and their combination to induce T‑cell activation. The quality and duration of the T‑cell response can be specifically modulated by modifying the modular composition of the CAR; CAR T‑cells can act as "biopharmaceutical factories" (T-cells redirected for unrestricted cytokine-mediated killing, TRUCK) in the tissue by CAR-mediated release of transgenic therapeutic proteins. CONCLUSION Adoptive CAR T‑cell therapy has shown clinical efficacy in the treatment of hematological malignancies; the treatment of solid tumors, however, is more challenging. Allogeneic CAR T‑cell technology is aimed at generating "off-the-shelf" CAR T‑cells that are accessible for a large number of patients. A further promising approach is the use of CAR T‑cells for other therapeutic applications such as the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- A Holzinger
- Abteilung für Gen-Immuntherapie, RCI Regensburger Centrum für Interventionelle Immunologie, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland.,Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Deutschland
| | - H Abken
- Abteilung für Gen-Immuntherapie, RCI Regensburger Centrum für Interventionelle Immunologie, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Deutschland. .,Universitätsklinikum Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Deutschland.
| |
Collapse
|
110
|
Hypoxia-sensing CAR T cells provide safety and efficacy in treating solid tumors. CELL REPORTS MEDICINE 2021; 2:100227. [PMID: 33948568 PMCID: PMC8080111 DOI: 10.1016/j.xcrm.2021.100227] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/25/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
Utilizing T cells expressing chimeric antigen receptors (CARs) to identify and attack solid tumors has proven challenging, in large part because of the lack of tumor-specific targets to direct CAR binding. Tumor selectivity is crucial because on-target, off-tumor activation of CAR T cells can result in potentially lethal toxicities. This study presents a stringent hypoxia-sensing CAR T cell system that achieves selective expression of a pan-ErbB-targeted CAR within a solid tumor, a microenvironment characterized by inadequate oxygen supply. Using murine xenograft models, we demonstrate that, despite widespread expression of ErbB receptors in healthy organs, the approach provides anti-tumor efficacy without off-tumor toxicity. This dynamic on/off oxygen-sensing safety switch has the potential to facilitate unlimited expansion of the CAR T cell target repertoire for treating solid malignancies. A dual oxygen-sensing switch provides stringent hypoxia-dependent regulation of a CAR HypoxiCAR T cells deliver tumor-selective CAR expression and anti-tumor efficacy HypoxiCAR T cells prevent on-target, off-tumor activation and cytokine release syndrome HypoxiCAR provides a strategy to expand the CAR repertoire for solid malignancies
Collapse
|
111
|
CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J 2021; 11:69. [PMID: 33824268 PMCID: PMC8024391 DOI: 10.1038/s41408-021-00459-7] [Citation(s) in RCA: 1087] [Impact Index Per Article: 362.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 02/01/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is a revolutionary new pillar in cancer treatment. Although treatment with CAR-T cells has produced remarkable clinical responses with certain subsets of B cell leukemia or lymphoma, many challenges limit the therapeutic efficacy of CAR-T cells in solid tumors and hematological malignancies. Barriers to effective CAR-T cell therapy include severe life-threatening toxicities, modest anti-tumor activity, antigen escape, restricted trafficking, and limited tumor infiltration. In addition, the host and tumor microenvironment interactions with CAR-T cells critically alter CAR-T cell function. Furthermore, a complex workforce is required to develop and implement these treatments. In order to overcome these significant challenges, innovative strategies and approaches to engineer more powerful CAR-T cells with improved anti-tumor activity and decreased toxicity are necessary. In this review, we discuss recent innovations in CAR-T cell engineering to improve clinical efficacy in both hematological malignancy and solid tumors and strategies to overcome limitations of CAR-T cell therapy in both hematological malignancy and solid tumors.
Collapse
|
112
|
Schwerdtfeger M, Benmebarek MR, Endres S, Subklewe M, Desiderio V, Kobold S. Chimeric Antigen Receptor-Modified T Cells and T Cell-Engaging Bispecific Antibodies: Different Tools for the Same Job. Curr Hematol Malig Rep 2021; 16:218-233. [PMID: 33939108 PMCID: PMC8154758 DOI: 10.1007/s11899-021-00628-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Both chimeric antigen receptor (CAR) T cells and T cell-engaging antibodies (BiAb) have been approved for the treatment of hematological malignancies. However, despite targeting the same antigen, they represent very different classes of therapeutics, each with its distinct advantages and drawbacks. In this review, we compare BiAb and CAR T cells with regard to their mechanism of action, manufacturing, and clinical application. In addition, we present novel strategies to overcome limitations of either approach and to combine the best of both worlds. RECENT FINDINGS By now there are multiple approaches combining the advantages of BiAb and CAR T cells. A major area of research is the application of both formats for solid tumor entities. This includes improving the infiltration of T cells into the tumor, counteracting immunosuppression in the tumor microenvironment, targeting antigen heterogeneity, and limiting off-tumor on-target effects. BiAb come with the major advantage of being an off-the-shelf product and are more controllable because of their half-life. They have also been reported to induce less frequent and less severe adverse events. CAR T cells in turn demonstrate superior response rates, have the potential for long-term persistence, and can be additionally genetically modified to overcome some of their limitations, e.g., to make them more controllable.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/genetics
- Antibodies, Bispecific/immunology
- Antigens, Neoplasm/immunology
- Genetic Engineering
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Neoplasms/etiology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Signal Transduction
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Melanie Schwerdtfeger
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Munich, Germany
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Munich, Germany
- German Center for Translational Cancer Research (DKTK), Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Marion Subklewe
- Department of Medicine III, Klinikum der Universität München, LMU Munich, Munich, Germany
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Munich, Germany
- German Center for Translational Cancer Research (DKTK), Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| |
Collapse
|
113
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Rahbarizadeh F, Khoshtinat Nikkhoi S. Strategies for Dodging the Obstacles in CAR T Cell Therapy. Front Oncol 2021; 11:627549. [PMID: 33869011 PMCID: PMC8047470 DOI: 10.3389/fonc.2021.627549] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has offered cancer patients a new alternative therapeutic choice in recent years. This novel type of therapy holds tremendous promise for the treatment of various hematologic malignancies including B-cell acute lymphoblastic leukemia (B-ALL) and lymphoma. However, CAR T cell therapy has experienced its ups and downs in terms of toxicities and efficacy shortcomings. Adverse events such as cytokine release syndrome (CRS), neurotoxicity, graft rejection, on-target off-tumor toxicities, and tumor relapse have tied the rescuing hands of CAR T cell therapies. Moreover, in the case of solid tumor treatment, CAR T cell therapies have not yielded encouraging results mainly due to challenges such as the formidable network of the tumor microenvironments (TME) that operates in a suppressive fashion resulting in CAR T cell dysfunction. In this review, we tend to shine a light on emerging strategies and solutions for addressing the mentioned barriers. These solutions might dramatically help shorten the gap between a successful clinical outcome and the hope for it.
Collapse
Affiliation(s)
- Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.,Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
114
|
Marofi F, Tahmasebi S, Rahman HS, Kaigorodov D, Markov A, Yumashev AV, Shomali N, Chartrand MS, Pathak Y, Mohammed RN, Jarahian M, Motavalli R, Motavalli Khiavi F. Any closer to successful therapy of multiple myeloma? CAR-T cell is a good reason for optimism. Stem Cell Res Ther 2021; 12:217. [PMID: 33781320 PMCID: PMC8008571 DOI: 10.1186/s13287-021-02283-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022] Open
Abstract
Despite many recent advances on cancer novel therapies, researchers have yet a long way to cure cancer. They have to deal with tough challenges before they can reach success. Nonetheless, it seems that recently developed immunotherapy-based therapy approaches such as adoptive cell transfer (ACT) have emerged as a promising therapeutic strategy against various kinds of tumors even the cancers in the blood (liquid cancers). The hematological (liquid) cancers are hard to be targeted by usual cancer therapies, for they do not form localized solid tumors. Until recently, two types of ACTs have been developed and introduced; tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR)-T cells which the latter is the subject of our discussion. It is interesting about engineered CAR-T cells that they are genetically endowed with unique cancer-specific characteristics, so they can use the potency of the host immune system to fight against either solid or liquid cancers. Multiple myeloma (MM) or simply referred to as myeloma is a type of hematological malignancy that affects the plasma cells. The cancerous plasma cells produce immunoglobulins (antibodies) uncontrollably which consequently damage the tissues and organs and break the immune system function. Although the last few years have seen significant progressions in the treatment of MM, still a complete remission remains unconvincing. MM is a medically challenging and stubborn disease with a disappointingly low rate of survival rate. When comparing the three most occurring blood cancers (i.e., lymphoma, leukemia, and myeloma), myeloma has the lowest 5-year survival rate (around 40%). A low survival rate indicates a high mortality rate with difficulty in treatment. Therefore, novel CAR-T cell-based therapies or combination therapies along with CAT-T cells may bring new hope for multiple myeloma patients. CAR-T cell therapy has a high potential to improve the remission success rate in patients with MM. To date, many preclinical and clinical trial studies have been conducted to investigate the ability and capacity of CAR T cells in targeting the antigens on myeloma cells. Despite the problems and obstacles, CAR-T cell experiments in MM patients revealed a robust therapeutic potential. However, several factors might be considered during CAR-T cell therapy for better response and reduced side effects. Also, incorporating the CAT-T cell method into a combinational treatment schedule may be a promising approach. In this paper, with a greater emphasis on CAR-T cell application in the treatment of MM, we will discuss and introduce CAR-T cell's history and functions, their limitations, and the solutions to defeat the limitations and different types of modifications on CAR-T cells.
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safa Tahmasebi
- Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Sulaymaniyah, Iraq
| | - Denis Kaigorodov
- Director of Research Institute "MitoKey", Moscow State Medical University, Moscow, Russian Federation
| | | | - Alexei Valerievich Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Trubetskaya St., 8-2, Moscow, Russian Federation, 119991
| | - Navid Shomali
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Yashwant Pathak
- Faculty Affairs, Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA.,Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Rebar N Mohammed
- Bone Marrow Transplant Center, Hiwa Cancer Hospital, Suleimanyah, Iraq
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120, Heidelberg, Germany
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | |
Collapse
|
115
|
Akhoundi M, Mohammadi M, Sahraei SS, Sheykhhasan M, Fayazi N. CAR T cell therapy as a promising approach in cancer immunotherapy: challenges and opportunities. Cell Oncol (Dordr) 2021; 44:495-523. [PMID: 33759063 DOI: 10.1007/s13402-021-00593-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-modified T cell therapy has shown great potential in the immunotherapy of patients with hematologic malignancies. In spite of this striking achievement, there are still major challenges to overcome in CAR T cell therapy of solid tumors, including treatment-related toxicity and specificity. Also, other obstacles may be encountered in tackling solid tumors, such as their immunosuppressive microenvironment, the heterogeneous expression of cell surface markers, and the cumbersome arrival of T cells at the tumor site. Although several strategies have been developed to overcome these challenges, aditional research aimed at enhancing its efficacy with minimum side effects, the design of precise yet simplified work flows and the possibility to scale-up production with reduced costs and related risks is still warranted. CONCLUSIONS Here, we review main strategies to establish a balance between the toxicity and activity of CAR T cells in order to enhance their specificity and surpass immunosuppression. In recent years, many clinical studies have been conducted that eventually led to approved products. To date, the FDA has approved two anti-CD19 CAR T cell products for non-Hodgkin lymphoma therapy, i.e., axicbtagene ciloleucel and tisagenlecleucel. With all the advances that have been made in the field of CAR T cell therapy for hematologic malignancies therapy, ongoing studies are focused on optimizing its efficacy and specificity, as well as reducing the side effects. Also, the efforts are poised to broaden CAR T cell therapeutics for other cancers, especially solid tumors.
Collapse
Affiliation(s)
- Maryam Akhoundi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh Saeideh Sahraei
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
116
|
Marofi F, Rahman HS, Thangavelu L, Dorofeev A, Bayas-Morejón F, Shirafkan N, Shomali N, Chartrand MS, Jarahian M, Vahedi G, Mohammed RN, Shahrokh S, Akbari M, Khiavi FM. Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy. Stem Cell Res Ther 2021; 12:200. [PMID: 33752707 PMCID: PMC7983395 DOI: 10.1186/s13287-021-02251-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/28/2021] [Indexed: 02/13/2023] Open
Abstract
In recent decades, a new method of cellular immunotherapy was introduced based on engineering and empowering the immune effector cells. In this type of immunotherapy, the immune effector cells are equipped with chimeric antigen receptor (CAR) to specifically target cancer cells. In much of the trials and experiments, CAR-modified T cell immunotherapy has achieved very promising therapeutic results in the treatment of some types of cancers and infectious diseases. However, there are also some considerable drawbacks in the clinical application of CAR-T cells although much effort is in progress to rectify the issues. In some conditions, CAR-T cells initiate over-activated and strong immune responses, therefore, causing unexpected side-effects such as systemic cytokine toxicity (i.e., cytokine release syndrome), neurotoxicity, on-target, off-tumor toxicity, and graft-versus-host disease (GvHD). To overcome these limitations in CAR-T cell immunotherapy, NK cells as an alternative source of immune effector cells have been utilized for CAR-engineering. Natural killer cells are key players of the innate immune system that can destroy virus-infected cells, tumor cells, or other aberrant cells with their efficient recognizing capability. Compared to T cells, CAR-transduced NK cells (CAR-NK) have several advantages, such as safety in clinical use, non-MHC-restricted recognition of tumor cells, and renewable and easy cell sources for their preparation. In this review, we will discuss the recent preclinical and clinical studies, different sources of NK cells, transduction methods, possible limitations and challenges, and clinical considerations.
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Sulaymaniyah, Iraq
| | - Lakshmi Thangavelu
- Associate professor, Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Aleksey Dorofeev
- Department of Propaedeutics of Dental Diseases, I.M. Sechenov First Moscow State Medical University (Sechenov University,), Moscow, Russian Federation
| | - Favian Bayas-Morejón
- Center for Research and Biotechnological Development, Research Department, Bolivar State University, Faculty of Agricultural Sciences, Natural Resources and the Environment, CP 020150 Guaranda, Ecuador
| | - Naghmeh Shirafkan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), 69120 Heidelberg, Germany
| | - Ghasem Vahedi
- Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Rebar N. Mohammed
- College of Veterinary Medicine, University of Sulaimani, Suleimanyah, Iraq
| | - Somayeh Shahrokh
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Shahrekord, Shahrekord, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
117
|
Schäfer D, Tomiuk S, Küster LN, Rawashdeh WA, Henze J, Tischler-Höhle G, Agorku DJ, Brauner J, Linnartz C, Lock D, Kaiser A, Herbel C, Eckardt D, Lamorte M, Lenhard D, Schüler J, Ströbel P, Missbach-Guentner J, Pinkert-Leetsch D, Alves F, Bosio A, Hardt O. Identification of CD318, TSPAN8 and CD66c as target candidates for CAR T cell based immunotherapy of pancreatic adenocarcinoma. Nat Commun 2021; 12:1453. [PMID: 33674603 PMCID: PMC7935963 DOI: 10.1038/s41467-021-21774-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
A major roadblock prohibiting effective cellular immunotherapy of pancreatic ductal adenocarcinoma (PDAC) is the lack of suitable tumor-specific antigens. To address this challenge, here we combine flow cytometry screenings, bioinformatic expression analyses and a cyclic immunofluorescence platform. We identify CLA, CD66c, CD318 and TSPAN8 as target candidates among 371 antigens and generate 32 CARs specific for these molecules. CAR T cell activity is evaluated in vitro based on target cell lysis, T cell activation and cytokine release. Promising constructs are evaluated in vivo. CAR T cells specific for CD66c, CD318 and TSPAN8 demonstrate efficacies ranging from stabilized disease to complete tumor eradication with CD318 followed by TSPAN8 being the most promising candidates for clinical translation based on functionality and predicted safety profiles. This study reveals potential target candidates for CAR T cell based immunotherapy of PDAC together with a functional set of CAR constructs specific for these molecules.
Collapse
Affiliation(s)
- Daniel Schäfer
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
- University Medical Center Göttingen, Clinic for Hematology and Medical Oncology, Göttingen, Lower Saxony, Germany
- University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology, Göttingen, Lower Saxony, Germany
| | - Stefan Tomiuk
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Laura N Küster
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Wa'el Al Rawashdeh
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Janina Henze
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
- University Medical Center Göttingen, Clinic for Hematology and Medical Oncology, Göttingen, Lower Saxony, Germany
- University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology, Göttingen, Lower Saxony, Germany
| | | | - David J Agorku
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Janina Brauner
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Cathrin Linnartz
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Dominik Lock
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Andrew Kaiser
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Christoph Herbel
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Dominik Eckardt
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Melina Lamorte
- Charles River Discovery Research Services GmbH, Freiburg, Baden-Wuerttemberg, Germany
| | - Dorothee Lenhard
- Charles River Discovery Research Services GmbH, Freiburg, Baden-Wuerttemberg, Germany
| | - Julia Schüler
- Charles River Discovery Research Services GmbH, Freiburg, Baden-Wuerttemberg, Germany
| | - Philipp Ströbel
- University Medical Center Göttingen, Institute for Pathology, Göttingen, Lower Saxony, Germany
| | - Jeannine Missbach-Guentner
- University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology, Göttingen, Lower Saxony, Germany
| | - Diana Pinkert-Leetsch
- University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology, Göttingen, Lower Saxony, Germany
- Max Planck Institute for Experimental Medicine, Translational Molecular Imaging, Göttingen, Lower Saxony, Germany
| | - Frauke Alves
- University Medical Center Göttingen, Clinic for Hematology and Medical Oncology, Göttingen, Lower Saxony, Germany
- University Medical Center Göttingen, Institute for Diagnostic and Interventional Radiology, Göttingen, Lower Saxony, Germany
- Max Planck Institute for Experimental Medicine, Translational Molecular Imaging, Göttingen, Lower Saxony, Germany
| | - Andreas Bosio
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany
| | - Olaf Hardt
- Miltenyi Biotec GmbH, R&D, Bergisch Gladbach, North Rhine-Westphalia, Germany.
| |
Collapse
|
118
|
Martínez Bedoya D, Dutoit V, Migliorini D. Allogeneic CAR T Cells: An Alternative to Overcome Challenges of CAR T Cell Therapy in Glioblastoma. Front Immunol 2021; 12:640082. [PMID: 33746981 PMCID: PMC7966522 DOI: 10.3389/fimmu.2021.640082] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as one of the major breakthroughs in cancer immunotherapy in the last decade. Outstanding results in hematological malignancies and encouraging pre-clinical anti-tumor activity against a wide range of solid tumors have made CAR T cells one of the most promising fields for cancer therapies. CAR T cell therapy is currently being investigated in solid tumors including glioblastoma (GBM), a tumor for which survival has only modestly improved over the past decades. CAR T cells targeting EGFRvIII, Her2, or IL-13Rα2 have been tested in GBM, but the first clinical trials have shown modest results, potentially due to GBM heterogeneity and to the presence of an immunosuppressive microenvironment. Until now, the use of autologous T cells to manufacture CAR products has been the norm, but this approach has several disadvantages regarding production time, cost, manufacturing delay and dependence on functional fitness of patient T cells, often reduced by the disease or previous therapies. Universal “off-the-shelf,” or allogeneic, CAR T cells is an alternative that can potentially overcome these issues, and allow for multiple modifications and CAR combinations to target multiple tumor antigens and avoid tumor escape. Advances in genome editing tools, especially via CRISPR/Cas9, might allow overcoming the two main limitations of allogeneic CAR T cells product, i.e., graft-vs.-host disease and host allorejection. Here, we will discuss how allogeneic CAR T cells could allow for multivalent approaches and alteration of the tumor microenvironment, potentially allowing the development of next generation therapies for the treatment of patients with GBM.
Collapse
Affiliation(s)
- Darel Martínez Bedoya
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.,Swiss Cancer Center Léman, Lausanne, Switzerland.,Brain Tumor and Immune Cell Engineering Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Valérie Dutoit
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.,Swiss Cancer Center Léman, Lausanne, Switzerland.,Brain Tumor and Immune Cell Engineering Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Denis Migliorini
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.,Swiss Cancer Center Léman, Lausanne, Switzerland.,Brain Tumor and Immune Cell Engineering Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
119
|
Schoenfeld AJ, O'Cearbhaill RE. How Do We Meet the Challenge of Chimeric Antigen Receptor T-Cell Therapy for Solid Tumors? Cancer J 2021; 27:134-142. [PMID: 33750073 PMCID: PMC8457037 DOI: 10.1097/ppo.0000000000000516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
ABSTRACT Immune checkpoint inhibition has vastly improved the treatment of solid tumors, but most patients do not experience durable clinical benefit, so novel immunotherapeutic approaches are needed. Autologous T cells genetically engineered to express chimeric antigen receptors (CARs) have led to unprecedented clinical success in hematologic malignancies, and increasing efforts are actively being pursued to translate these benefits to the solid tumor arena. However, solid tumors present unique challenges for CAR T-cell development. In this review, we examine the potential barriers to progress and present emerging approaches to overcome these challenges with CAR therapy in solid tumors.
Collapse
Affiliation(s)
- Adam J Schoenfeld
- From the Cellular Therapy Center, Department of Medicine, Memorial Sloan Kettering Cancer Center, and Department of Medicine, Weill Cornell Medical College, New York, NY
| | | |
Collapse
|
120
|
Li R, An Y, Jin T, Zhang F, He P. Detection of MUC1 protein on tumor cells and their derived exosomes for breast cancer surveillance with an electrochemiluminescence aptasensor. J Electroanal Chem (Lausanne) 2021. [DOI: 10.1016/j.jelechem.2021.115011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
121
|
Abstract
Cancer is a major burden on the healthcare system, and new therapies are needed. Recently, the development of immunotherapies, which aim to boost or use the immune system, or its constituents, as a tool to fight malignant cells, has provided a major new tool in the arsenal of clinicians and has revolutionized the treatment of many cancers.Cellular immunotherapies are based on the administration of living cells to patients and have developed hugely, especially since 2010 when Sipuleucel-T (Provenge), a DC vaccine, was the first cellular immunotherapy to be approved by the FDA. The ensuing years have seen two further cellular immunotherapies gain FDA approval: tisagenlecleucel (Kymriah) and axicabtagene ciloleucel (Yescarta).This review will give an overview of the principles of immunotherapies before focusing on the major forms of cellular immunotherapies individually, T cell-based, natural killer (NK) cell-based and dendritic cell (DC)-based, as well as detailing some of the clinical trials relevant to each therapy.
Collapse
Affiliation(s)
- Conall Hayes
- School of Medicine, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
122
|
Greenshpan Y, Sharabi O, Ottolenghi A, Cahana A, Kundu K, M Yegodayev K, Elkabets M, Gazit R, Porgador A. Synthetic promoters to induce immune-effectors into the tumor microenvironment. Commun Biol 2021; 4:143. [PMID: 33514819 PMCID: PMC7846768 DOI: 10.1038/s42003-021-01664-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 11/20/2020] [Indexed: 11/21/2022] Open
Abstract
Harnessing the immune-system to eradicate cancer is becoming a reality in recent years. Engineered immune cells, such as chimeric antigen receptor (CAR) T cells, are facing the danger of an overt life-threatening immune response due to the ON-target OFF-tumor cytotoxicity and Cytokine Release Syndrome. We therefore developed synthetic promoters for regulation of gene expression under the control of inflammation and Hypoxia-induced signals that are associated with the tumor microenvironment (TME). We termed this methodology as chimeric-antigen-receptor-tumor-induced-vector (CARTIV). For proof of concept, we studied synthetic promoters based on promoter-responsive elements (PREs) of IFNγ, TNFα and hypoxia; triple PRE-based CARTIV promoter manifested a synergistic activity in cell-lines and potent activation in human primary T-cells. CARTIV platform can improve safety of CAR T-cells or other engineered immune-cells, providing TME-focused activity and opening a therapeutic window for many tumor-associated antigens that are also expressed by non-tumor healthy tissues.
Collapse
Affiliation(s)
- Yariv Greenshpan
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omri Sharabi
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aner Ottolenghi
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Avishag Cahana
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kiran Kundu
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ksenia M Yegodayev
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Moshe Elkabets
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Roi Gazit
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Angel Porgador
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
123
|
Li H, Yang C, Cheng H, Huang S, Zheng Y. CAR-T cells for Colorectal Cancer: Target-selection and strategies for improved activity and safety. J Cancer 2021; 12:1804-1814. [PMID: 33613769 PMCID: PMC7890323 DOI: 10.7150/jca.50509] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/29/2020] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cell immunotherapy is a novel method that is genetically engineered to recruit T cells against malignant disease. Administration of CAR-T cells has led to progress in hematological malignancies, and it has been proposed for solid tumors like colorectal cancer (CRC) for years. However, this method was not living up to expectations for the intrinsic challenges posed to CAR-T cells by solid tumors, which mainly due to the lacking of tumor-restricted antigens and adverse effects following treatment. New approaches are proposed to overcome the multiple challenges to alleviate the difficult situation of CAR-T cells in CRC, including engineering T cells with immune-activating molecules, regional administration of T cell, bispecific T cell engager, and combinatorial target-antigen recognition. In this review, we sum up the current stage of knowledge about target-selection, adverse events like on/off-tumor toxicity, the preclinical and clinical studies of CAR-T therapy, and the characteristics of strategies applied in CRC.
Collapse
Affiliation(s)
- Huali Li
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chao Yang
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Huangrong Cheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shuoyang Huang
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
124
|
Shu R, Evtimov VJ, Hammett MV, Nguyen NYN, Zhuang J, Hudson PJ, Howard MC, Pupovac A, Trounson AO, Boyd RL. Engineered CAR-T cells targeting TAG-72 and CD47 in ovarian cancer. MOLECULAR THERAPY-ONCOLYTICS 2021; 20:325-341. [PMID: 33614914 PMCID: PMC7868933 DOI: 10.1016/j.omto.2021.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/09/2021] [Indexed: 02/04/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have revolutionized blood cancer immunotherapy; however, their efficacy against solid tumors has been limited. A common mechanism of tumor escape from single target therapies is downregulation or mutational loss of the nominal epitope. Targeting multiple antigens may thus improve the effectiveness of CAR immunotherapies. We generated dual CAR-T cells targeting two tumor antigens: TAG-72 (tumor-associated glycoprotein 72) and CD47. TAG-72 is a pan-adenocarcinoma oncofetal antigen, highly expressed in ovarian cancers, with increased expression linked to disease progression. CD47 is ubiquitously overexpressed in multiple tumor types, including ovarian cancer; it is a macrophage “don’t eat me” signal. However, CD47 is also expressed on many normal cells. To avoid this component of the dual CAR-T cells killing healthy tissue, we designed a truncated CD47 CAR devoid of intracellular signaling domains. The CD47 CAR facilitates binding to CD47+ cells, increasing the prospect of TAG-72+ cell elimination via the TAG-72 CAR. Furthermore, we could reduce the damage to normal tissue by monomerizing the CD47 CAR. Our results indicate that the co-expression of the TAG-72 CAR and the CD47-truncated monomer CAR on T cells could be an effective, dual CAR-T cell strategy for ovarian cancer, also applicable to other adenocarcinomas.
Collapse
Affiliation(s)
- Runzhe Shu
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia
| | | | | | | | - Junli Zhuang
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia
| | - Peter J Hudson
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia.,Avipep Pty, Ltd., Parkville, VIC 3052, Australia
| | | | | | - Alan O Trounson
- Cartherics Pty, Ltd., Clayton, VIC 3168, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | | |
Collapse
|
125
|
Hematopoietic versus Solid Cancers and T Cell Dysfunction: Looking for Similarities and Distinctions. Cancers (Basel) 2021; 13:cancers13020284. [PMID: 33466674 PMCID: PMC7828769 DOI: 10.3390/cancers13020284] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Dysfunction of the immune T cell compartment occurs in many hematopoietic as well as solid cancers and hampers successful application of new immunotherapeutic approaches. A complete understanding of T cell dysfunction might improve the outcome of such therapies, but an overview in the various cancers is still lacking. We aim to map areas of similarities and differences in solid versus hematopoietic malignancies, providing a high-level rather than a detailed perspective on T cell dysfunction in those tumors. Abstract Cancer cells escape, suppress and exploit the host immune system to sustain themselves, and the tumor microenvironment (TME) actively dampens T cell function by various mechanisms. Over the last years, new immunotherapeutic approaches, such as adoptive chimeric antigen receptor (CAR) T cell therapy and immune checkpoint inhibitors, have been successfully applied for refractory malignancies that could only be treated in a palliative manner previously. Engaging the anti-tumor activity of the immune system, including CAR T cell therapy to target the CD19 B cell antigen, proved to be effective in acute lymphocytic leukemia. In low-grade hematopoietic B cell malignancies, such as chronic lymphocytic leukemia, clinical outcomes have been tempered by cancer-induced T cell dysfunction characterized in part by a state of metabolic lethargy. In multiple myeloma, novel antigens such as BCMA and CD38 are being explored for CAR T cells. In solid cancers, T cell-based immunotherapies have been applied successfully to melanoma and lung cancers, whereas application in e.g., breast cancer lags behind and is modestly effective as yet. The main hurdles for CAR T cell immunotherapy in solid tumors are the lack of suitable antigens, anatomical inaccessibility, and T cell anergy due to immunosuppressive TME. Given the wide range of success and failure of immunotherapies in various cancer types, it is crucial to comprehend the underlying similarities and distinctions in T cell dysfunction. Hence, this review aims at comparing selected, distinct B cell-derived versus solid cancer types and at describing means by which malignant cells and TME might dampen T cell anti-tumor activity, with special focus on immunometabolism. Drawing a meaningful parallel between the efficacy of immunotherapy and the extent of T cell dysfunction will shed light on areas where we can improve immune function to battle cancer.
Collapse
|
126
|
Abstract
Adoptive cellular therapy (ACT) is a form of cancer immunotherapy in which lymphocytes are removed from patient blood or tumor samples, expanded and/or genetically modified to improve tumor-fighting capabilities, and infused back into the patient. The main forms of ACT include tumor infiltrating lymphocytes (TILs), engineered T cell receptor (TCR) T cells, and chimeric antigen receptor (CAR) T cells. While ACT has had success in hematological malignancies, particularly in B cell lymphomas targeted with CAR T cells, these favorable outcomes have yet to be replicated in solid tumors. Appropriate solid tumor target antigens are difficult to identify for ACT. Trafficking to tumor sites and infiltrating solid tumor burdens remains a problem for ACT cells. Persistence of ACT cells, which is important in creating a durable response, is also a major challenge, partly attributed to the formidable microtumor environment conditions. The costly and time-intensive manufacturing process for ACT is also an obstacle to widespread adoption. In this review, we discuss the challenges of ACT therapy in the treatment of solid tumors and explore the ongoing efforts to improve this immunotherapy approach in non-hematological malignancies.
Collapse
Affiliation(s)
- Joseph M Grimes
- Columbia University Vagelos College of Physicians and Surgeons, 630 W. 168th St., New York, NY, 10032, United States.
| | - Richard D Carvajal
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 177 Fort Washington Avenue, New York, NY, 10032, United States.
| | - Pawel Muranski
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, United States.
| |
Collapse
|
127
|
Cortés-Hernández A, Alvarez-Salazar EK, Soldevila G. Chimeric Antigen Receptor (CAR) T Cell Therapy for Cancer. Challenges and Opportunities: An Overview. Methods Mol Biol 2021; 2174:219-244. [PMID: 32813253 DOI: 10.1007/978-1-0716-0759-6_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The use of immunotherapy as an alternative treatment for cancer patients has become of great interest in the scientific community as it is required to overcome many of the currently unsolved problems such as tumor escape, immunosuppression and unwanted unspecific toxicity. The use of chimeric antigen receptor T cells has been a very successful strategy in some hematologic malignancies. However, the application of CAR T cells has been limited to solid tumors, and this has aimed the development of new generation of CARs with enhanced effectivity and specificity. Here, we review the state of the art of CAR T cell therapy with special emphasis on the current challenges and opportunities.
Collapse
Affiliation(s)
- Arimelek Cortés-Hernández
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Evelyn Katy Alvarez-Salazar
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México.
| |
Collapse
|
128
|
Bonfá G, Blazquez-Roman J, Tarnai R, Siciliano V. Precision Tools in Immuno-Oncology: Synthetic Gene Circuits for Cancer Immunotherapy. Vaccines (Basel) 2020; 8:E732. [PMID: 33287392 PMCID: PMC7761833 DOI: 10.3390/vaccines8040732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
Engineered mammalian cells for medical purposes are becoming a clinically relevant reality thanks to advances in synthetic biology that allow enhanced reliability and safety of cell-based therapies. However, their application is still hampered by challenges including time-consuming design-and-test cycle iterations and costs. For example, in the field of cancer immunotherapy, CAR-T cells targeting CD19 have already been clinically approved to treat several types of leukemia, but their use in the context of solid tumors is still quite inefficient, with additional issues related to the adequate quality control for clinical use. These limitations can be overtaken by innovative bioengineering approaches currently in development. Here we present an overview of recent synthetic biology strategies for mammalian cell therapies, with a special focus on the genetic engineering improvements on CAR-T cells, discussing scenarios for the next generation of genetic circuits for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Velia Siciliano
- Synthetic and Systems Biology Lab for Biomedicine, Istituto Italiano di Tecnologia-IIT, Largo Barsanti e Matteucci, 80125 Naples, Italy; (G.B.); (J.B.-R.); (R.T.)
| |
Collapse
|
129
|
A Functional Screening Strategy for Engineering Chimeric Antigen Receptors with Reduced On-Target, Off-Tumor Activation. Mol Ther 2020; 28:2564-2576. [PMID: 32827460 PMCID: PMC7704745 DOI: 10.1016/j.ymthe.2020.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/27/2020] [Accepted: 08/05/2020] [Indexed: 01/15/2023] Open
Abstract
In recent years, chimeric antigen receptor (CAR) T cell cancer immunotherapies have advanced substantially in the clinic. However, challenges related to safety persist; one major concern occurs when CARs trigger a response to antigen present on healthy cells (on-target, off-tumor response). A strategy to ameliorate this relies on the complex relationship between receptor affinity and signaling, such that one can engineer a CAR that is only activated by tumor cells expressing high antigen levels. Here, we developed a CAR T cell display platform with stable genomic expression and rapid functional screening based on interleukin-2 signaling. Starting with a CAR with high affinity toward its target antigen, we combined CRISPR-Cas9 genome editing and deep mutational scanning to generate a library of antigen-binding domain variants. This library was subjected to multiple rounds of selection based on either antigen binding or cell signaling. Deep sequencing of the resulting libraries and a comparative analysis revealed the enrichment and depletion of specific variants from which we selected CARs that were selectively activated by tumor cells based on antigen expression levels. Our platform demonstrates how directed evolution based on functional screening and deep sequencing-guided selection can be combined to enhance the selectivity and safety of CARs.
Collapse
|
130
|
Jin Y, Dong Y, Zhang J, Sun J, Liu Y, Chen Y. The toxicity of cell therapy: Mechanism, manifestations, and challenges. J Appl Toxicol 2020; 41:659-667. [DOI: 10.1002/jat.4100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Yongjia Jin
- Shanghai Electric Power Hospital Shanghai China
| | - Yan Dong
- Department of Musculoskeletal Oncology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology Shanghai Medical College, Fudan University Shanghai China
| | | | | | | | - Yong Chen
- Department of Musculoskeletal Oncology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology Shanghai Medical College, Fudan University Shanghai China
| |
Collapse
|
131
|
Xie M, Viviani M, Fussenegger M. Engineering precision therapies: lessons and motivations from the clinic. Synth Biol (Oxf) 2020; 6:ysaa024. [PMID: 33817342 PMCID: PMC7998714 DOI: 10.1093/synbio/ysaa024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
In the past decade, gene- and cell-based therapies have been at the forefront of the biomedical revolution. Synthetic biology, the engineering discipline of building sophisticated 'genetic software' to enable precise regulation of gene activities in living cells, has been a decisive success factor of these new therapies. Here, we discuss the core technologies and treatment strategies that have already gained approval for therapeutic applications in humans. We also review promising preclinical work that could either enhance the efficacy of existing treatment strategies or pave the way for new precision medicines to treat currently intractable human conditions.
Collapse
Affiliation(s)
- Mingqi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Mirta Viviani
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|
132
|
Andrea AE, Chiron A, Bessoles S, Hacein-Bey-Abina S. Engineering Next-Generation CAR-T Cells for Better Toxicity Management. Int J Mol Sci 2020; 21:E8620. [PMID: 33207607 PMCID: PMC7696189 DOI: 10.3390/ijms21228620] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Immunoadoptive therapy with genetically modified T lymphocytes expressing chimeric antigen receptors (CARs) has revolutionized the treatment of patients with hematologic cancers. Although clinical outcomes in B-cell malignancies are impressive, researchers are seeking to enhance the activity, persistence, and also safety of CAR-T cell therapy-notably with a view to mitigating potentially serious or even life-threatening adverse events like on-target/off-tumor toxicity and (in particular) cytokine release syndrome. A variety of safety strategies have been developed by replacing or adding various components (such as OFF- and ON-switch CARs) or by combining multi-antigen-targeting OR-, AND- and NOT-gate CAR-T cells. This research has laid the foundations for a whole new generation of therapeutic CAR-T cells. Here, we review the most promising CAR-T cell safety strategies and the corresponding preclinical and clinical studies.
Collapse
Affiliation(s)
- Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut 1100, Lebanon;
| | - Andrada Chiron
- Université de Paris, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France; (A.C.); (S.B.)
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, 94275 Le-Kremlin-Bicêtre, France
| | - Stéphanie Bessoles
- Université de Paris, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France; (A.C.); (S.B.)
| | - Salima Hacein-Bey-Abina
- Université de Paris, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, F-75006 Paris, France; (A.C.); (S.B.)
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, 94275 Le-Kremlin-Bicêtre, France
| |
Collapse
|
133
|
Liao Q, Mao Y, He H, Ding X, Zhang X, Xu J. PD-L1 chimeric costimulatory receptor improves the efficacy of CAR-T cells for PD-L1-positive solid tumors and reduces toxicity in vivo. Biomark Res 2020; 8:57. [PMID: 33292688 PMCID: PMC7607631 DOI: 10.1186/s40364-020-00237-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND On-target off-tumor toxicity impedes the clinical application of chimeric antigen receptor-modified T cells (CAR-T cells) in the treatment of solid tumors. Previous reports proved that the combinatorial antigen recognition strategy could improve the safety profile of CAR-T cells by targeting two different tumor-associated antigens (TAAs), one as a CAR-T targeted antigen and the other as a chimeric costimulatory receptor (CCR) ligand. The programmed death-ligand 1 (PD-L1, also known as B7-H1) is preferentially overexpressed on multiple tumors, it will be highly interesting to explore the potential of PD-L1 as a universal target for designing CCR. METHODS A novel dual-targeted CAR, which is composed of first-generation CD19/HER2 CAR with CD3ζ signaling domain and PD-L1 CCR containing the CD28 costimulatory domain, was constructed and delivered into T cells by pseudotyped lentivirus. The cytokine release, cytotoxicity and proliferation of dual-targeted CAR-T cells were tested in vitro, and their safety and therapeutic efficacy were evaluated using a human tumor xenograft mouse model in vivo. RESULTS The dual-targeted CAR-T cells exerted a similar cytotoxic activity against CD19/HER2+ tumor cells with or without PD-L1 in vitro, however, enhanced cytokine releases and improved proliferative capacity were only observed in the presence of both CD19/HER2 and PD-L1. Importantly, the dual-targeted CAR-T cells displayed no cytotoxicity against PD-L1+ cells alone in the absence of tumor antigen CD19/HER2. In addition, the dual-targeted CAR-T cells preferably destroyed tumor xenografts bearing both CD19/HER2 and PD-L1, but spared only antigen-positive tumor xenografts without PD-L1 in vivo. Furthermore, PD-L1 CCR also improved the antitumor efficacy of the low-affinity HER2 CAR-T cells against PD-L1+ tumors expressing high levels of HER2. CONCLUSION Our observations demonstrated that PD-L1 could be used as a universal target antigen for designing CCR, and the dual-targeted CAR-T cells equipped with PD-L1 CCR could be used to reduce the risk of on-target off-tumor toxicity while retaining their potent antitumor efficacy in the treatment of PD-L1+ solid tumors.
Collapse
Affiliation(s)
- Qibin Liao
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yunyu Mao
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Huan He
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiangqing Ding
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Jianqing Xu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
134
|
Watanabe N, McKenna MK, Rosewell Shaw A, Suzuki M. Clinical CAR-T Cell and Oncolytic Virotherapy for Cancer Treatment. Mol Ther 2020; 29:505-520. [PMID: 33130314 DOI: 10.1016/j.ymthe.2020.10.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has recently garnered success with the induction of clinical responses in tumors, which are traditionally associated with poor outcomes. Chimeric antigen receptor T (CAR-T) cells and oncolytic viruses (OVs) have emerged as promising cancer immunotherapy agents. Herein, we provide an overview of the current clinical status of CAR-T cell and OV therapies. While preclinical studies have demonstrated curative potential, the benefit of CAR-T cells and OVs as single-agent treatments remains limited to a subset of patients. Combinations of different targeted therapies may be required to achieve efficient, durable responses against heterogeneous tumors, as well as the microenvironment. Using a combinatorial approach to take advantage of the unique features of CAR-T cells and OVs with other treatments can produce additive therapeutic effects. This review also discusses ongoing clinical evaluations of these combination strategies for improved outcomes in treatment of resistant malignancies.
Collapse
Affiliation(s)
- Norihiro Watanabe
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Mary Kathryn McKenna
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Amanda Rosewell Shaw
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Masataka Suzuki
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
135
|
Gotti M, Defrancesco I, D'Angelo M, Basso S, Crotto L, Marinelli A, Maccalli C, Iaconianni V. Cancer Immunotherapy Using Chimeric Antigen Receptor Expressing T-Cells: Present and Future Needs of Clinical Cancer Centers. Front Immunol 2020; 11:565236. [PMID: 33193333 PMCID: PMC7662555 DOI: 10.3389/fimmu.2020.565236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/06/2020] [Indexed: 12/30/2022] Open
Abstract
Chimeric Antigen Receptor-T cells (CAR-T) are considered novel biological agents, designed to selectively attack cancer cells expressing specific antigens, with demonstrated clinical activity in patients affected with relapsed/refractory B-cell malignancies. In consideration of their complexity, the use of CAR-T requires dedicated clinical setting and health care practitioners with expertise in the selection, treatment, and management of toxicities and side effects. Such issue appears particularly important when contextualized in the rapid progress of CAR-T cell treatment, translating into a constant need of updating and evolution. Moreover, the clinical grade manufacturing of CAR-T cells is complex and implies articulated regulatory and organizational aspects. The main goal of this review is to summarize and provide an accurate analysis of the clinical, logistic, and regulatory requirements of CAR-T cell centers. Finally, we describe a new occupational figure called “CAR-T specialist” devoted to the establishment and coordination of the required facilities and regulatory landscape in the context of cancer centers.
Collapse
Affiliation(s)
- Manuel Gotti
- Division of Hematology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | | | - Mario D'Angelo
- Department of Onco-hematology, Cell and Gene Therapy, Ospedale Pediatrico Bambino Gesù IRCCS, Roma, Italy
| | - Sabrina Basso
- Pediatric Hematology/Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.,Cell Factory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Crotto
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Alfredo Marinelli
- Operative Unit (OU) Neuroncology, University Federico II, Napoli, Italy.,IRCCS Neuromed Istituto Neurologico Mediterraneo Pozzilli (INM), Pozzilli, Italy
| | - Cristina Maccalli
- Laboratory of Immune and Biological Therapy, Research Department, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
136
|
Xie Y, Hu Y, Zhou N, Yao C, Wu L, Liu L, Chen F. CAR T-cell therapy for triple-negative breast cancer: Where we are. Cancer Lett 2020; 491:121-131. [PMID: 32795486 DOI: 10.1016/j.canlet.2020.07.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most complex and challenging breast cancer subtype to treat, and chemotherapy remains the standard of care. Clinically, TNBC has a relatively high rate of recurrence and poor prognosis, which leads to a significant effort to discover novel strategies to treat patients with these tumors. Currently, chimeric antigen receptor (CAR) T cell-based immunotherapy redirects the patient's immune system directly to recognize and eradicate tumor-associated antigens (TAAs) expressing tumor cells being explored as a treatment for TNBC. A steadily increasing research in CAR T-cell therapy targeting different TAAs in TNBC has reported. In this review, we introduce the CAR technology and summarize the potential TAAs, available CARs, the antitumor activity, and the related toxicity of CARs currently under investigation for TNBC. We also highlight the potential strategies to prevent/reduce potential "on target, off tumor" toxicity induced by CAR T-cell therapy. This review will help to explore proper targets to expand further the CAR T-cell therapy for TNBCs in the clinic.
Collapse
Affiliation(s)
- Yuetao Xie
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Yi Hu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Nawu Zhou
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Cuicui Yao
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Lixin Wu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China
| | - Lin Liu
- Everest Medical Care, 2010 West Chester Pike, Havertown, PA, 19083, USA
| | - Fang Chen
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, Guangdong, 518038, China.
| |
Collapse
|
137
|
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: 3.5] [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
|
138
|
Hong M, Clubb JD, Chen YY. Engineering CAR-T Cells for Next-Generation Cancer Therapy. Cancer Cell 2020; 38:473-488. [PMID: 32735779 DOI: 10.1016/j.ccell.2020.07.005] [Citation(s) in RCA: 353] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
Abstract
T cells engineered to express chimeric antigen receptors (CARs) with tumor specificity have shown remarkable success in treating patients with hematologic malignancies and revitalized the field of adoptive cell therapy. However, realizing broader therapeutic applications of CAR-T cells necessitates engineering approaches on multiple levels to enhance efficacy and safety. Particularly, solid tumors present unique challenges due to the biological complexity of the solid-tumor microenvironment (TME). In this review, we highlight recent strategies to improve CAR-T cell therapy by engineering (1) the CAR protein, (2) T cells, and (3) the interaction between T cells and other components in the TME.
Collapse
Affiliation(s)
- Mihe Hong
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Justin D Clubb
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California-Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
139
|
Benard E, Casey NP, Inderberg EM, Wälchli S. SJI 2020 special issue: A catalogue of Ovarian Cancer targets for CAR therapy. Scand J Immunol 2020; 92:e12917. [PMID: 32557659 DOI: 10.1111/sji.12917] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Ovarian Cancer (OC) is currently difficult to cure, mainly due to its late detection and the advanced state of the disease at the time of diagnosis. Therefore, conventional treatments such as debulking surgery and combination chemotherapy are rarely able to control progression of the tumour, and relapses are frequent. Alternative therapies are currently being evaluated, including immunotherapy and advanced T cell-based therapy. In the present review, we will focus on a description of those Chimeric Antigen Receptors (CARs) that have been validated in the laboratory or are being tested in the clinic. Numerous target antigens have been defined due to the identification of OC biomarkers, and many are being used as CAR targets. We provide an exhaustive list of these constructs and their current status. Despite being innovative and efficient, the OC-specific CARs face a barrier to their clinical efficacy: the tumour microenvironment (TME). Indeed, effector cells expressing CARs have been shown to be severely inhibited, rendering the CAR T cells useless once at the tumour site. Herein, we give a thorough description of the highly immunosuppressive OC TME and present recent studies and innovations that have enabled CAR T cells to counteract this negative environment and to destroy tumours.
Collapse
Affiliation(s)
- Emmanuelle Benard
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Nicholas P Casey
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
140
|
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: 13] [Impact Index Per Article: 3.3] [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
|
141
|
Yang P, Wang Y, Yao Z, Gao X, Liu C, Wang X, Wu H, Ding X, Hu J, Lin B, Li Q, Li M, Li X, Chen X, Qi W, Li W, Xue J, Xu H. Enhanced Safety and Antitumor Efficacy of Switchable Dual Chimeric Antigen Receptor-Engineered T Cells against Solid Tumors through a Synthetic Bifunctional PD-L1-Blocking Peptide. J Am Chem Soc 2020; 142:18874-18885. [DOI: 10.1021/jacs.0c08538] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Peiwei Yang
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Ying Wang
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zheng Yao
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xinmei Gao
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Chen Liu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xinmin Wang
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Heming Wu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xu Ding
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jialiang Hu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Bingjing Lin
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Qian Li
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Mengwei Li
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xin Li
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xiangying Chen
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Weiyan Qi
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Weiguang Li
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jianpeng Xue
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Hanmei Xu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province and State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
142
|
Rafia C, Harly C, Scotet E. Beyond CAR T cells: Engineered Vγ9Vδ2 T cells to fight solid tumors. Immunol Rev 2020; 298:117-133. [DOI: 10.1111/imr.12920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Chirine Rafia
- INSERMCNRSCRCINAUniversité de Nantes Nantes France
- LabEx IGO “Immunotherapy, Graft, Oncology” Nantes France
- ImCheck Therapeutics Marseille France
| | - Christelle Harly
- INSERMCNRSCRCINAUniversité de Nantes Nantes France
- LabEx IGO “Immunotherapy, Graft, Oncology” Nantes France
| | - Emmanuel Scotet
- INSERMCNRSCRCINAUniversité de Nantes Nantes France
- LabEx IGO “Immunotherapy, Graft, Oncology” Nantes France
| |
Collapse
|
143
|
Date V, Nair S. Emerging vistas in CAR T-cell therapy: challenges and opportunities in solid tumors. Expert Opin Biol Ther 2020; 21:145-160. [PMID: 32882159 DOI: 10.1080/14712598.2020.1819978] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Despite advances in modern evidence-based medicine, cancer remains a major cause of global disease-associated mortality. CAR T-cell therapy is a major histocompatibility complex (MHC)-independent immunotherapy involving adoptive cell transfer. Cancer immunotherapy witnessed a major breakthrough with the US FDA approval of the first chimeric antigen receptor (CAR) T-cell therapy KymriahTM (tisagenlecleucel) for relapsed or refractory (R/R) acute lymphoblastic leukemia (ALL) in August 2017 followed by approval of Yescarta® (axicabtagene ciloleucel) for R/R non-Hodgkin's lymphoma (NHL) in October 2017. AREAS COVERED We review the potential of CAR T-cell therapy which, despite showing great promise in hematological malignancies, faces significant challenges in targeting solid tumors. We address these challenges and discuss proposed strategies to overcome them in solid tumors. We highlight the potential of CAR T-cell therapy as cancer precision medicine and briefly discuss the 'financial toxicity' of CAR T-cell therapy. EXPERT OPINION Taken together, we discuss various strategies to circumvent the limitations of CAR T-cell therapy in solid tumors. Despite the rapid advances in CAR NK-cell therapies, there is immense scope for CAR T-cell therapy in solid tumors. We provide a synthetic review of CAR T-cell therapy that will drive future research and harness its full potential in cancer precision medicine for solid tumors.
Collapse
Affiliation(s)
- Varada Date
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University , Mumbai, India
| | - Sujit Nair
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai , Mumbai, India
| |
Collapse
|
144
|
Editor's Pick: Tumour-Associated Hypoxia: Can We Give Chimeric Antigen Receptor T Cells More Breathing Space? EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR)-engineered T cells has encountered important limitations in the transition of their use from liquid to solid tumours. Success is dependent upon T-cell trafficking to, and functional persistence within, tumours that often present a metabolically and immunologically hostile microenvironment. Moreover, CAR targets that are tumour specific are extremely scarce. To address these issues, several strategies have been proposed to improve both tumour selectivity and safety. One approach involves the engineering of CAR-T cells that only deploy their effector function at tumour sites. Conceptually, a solution for this exploits the oxygen-limited nature of advanced tumour deposits through the engineering of CAR that are exclusively expressed or activated under conditions of profound hypoxia. T cells have a complex inter-relationship with oxygen, which also needs to be factored into the refinement of these technologies. Ideally, oxygen-sensing CAR should only function when oxygen tension is below 2%, as is commonly the case in solid tumours but rare in healthy tissue. Successful advancement of such technologies presents opportunities for solid tumour immunotherapy because it should broaden the target repertoire that may safely be exploited in this context.
Collapse
|
145
|
Wagner J, Wickman E, DeRenzo C, Gottschalk S. CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality? Mol Ther 2020; 28:2320-2339. [PMID: 32979309 DOI: 10.1016/j.ymthe.2020.09.015] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 01/07/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has garnered significant excitement due to its success for hematological malignancies in clinical studies leading to the US Food and Drug Administration (FDA) approval of three CD19-targeted CAR T cell products. In contrast, the clinical experience with CAR T cell therapy for solid tumors and brain tumors has been less encouraging, with only a few patients achieving complete responses. Clinical and preclinical studies have identified multiple "roadblocks," including (1) a limited array of targetable antigens and heterogeneous antigen expression, (2) limited T cell fitness and survival before reaching tumor sites, (3) an inability of T cells to efficiently traffic to tumor sites and penetrate physical barriers, and (4) an immunosuppressive tumor microenvironment. Herein, we review these challenges and discuss strategies that investigators have taken to improve the effector function of CAR T cells for the adoptive immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Jessica Wagner
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elizabeth Wickman
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher DeRenzo
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
146
|
Dannenfelser R, Allen GM, VanderSluis B, Koegel AK, Levinson S, Stark SR, Yao V, Tadych A, Troyanskaya OG, Lim WA. Discriminatory Power of Combinatorial Antigen Recognition in Cancer T Cell Therapies. Cell Syst 2020; 11:215-228.e5. [PMID: 32916097 DOI: 10.1016/j.cels.2020.08.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 06/08/2020] [Accepted: 08/04/2020] [Indexed: 12/23/2022]
Abstract
Precise discrimination of tumor from normal tissues remains a major roadblock for therapeutic efficacy of chimeric antigen receptor (CAR) T cells. Here, we perform a comprehensive in silico screen to identify multi-antigen signatures that improve tumor discrimination by CAR T cells engineered to integrate multiple antigen inputs via Boolean logic, e.g., AND and NOT. We screen >2.5 million dual antigens and ∼60 million triple antigens across 33 tumor types and 34 normal tissues. We find that dual antigens significantly outperform the best single clinically investigated CAR targets and confirm key predictions experimentally. Further, we identify antigen triplets that are predicted to show close to ideal tumor-versus-normal tissue discrimination for several tumor types. This work demonstrates the potential of 2- to 3-antigen Boolean logic gates for improving tumor discrimination by CAR T cell therapies. Our predictions are available on an interactive web server resource (antigen.princeton.edu).
Collapse
Affiliation(s)
- Ruth Dannenfelser
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Gregory M Allen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94158, USA; Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Ashley K Koegel
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94158, USA; Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; Division of Pediatric Oncology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sarah Levinson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sierra R Stark
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94158, USA; Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Vicky Yao
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Computer Science, Rice University, Houston, TX 77005, USA
| | - Alicja Tadych
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Olga G Troyanskaya
- Department of Computer Science, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA.
| | - Wendell A Lim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94158, USA; Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
147
|
Antoine P, Maher J. Developing a safe and effective CAR T-cell immunotherapy for breast cancer: progress and pitfalls. BREAST CANCER MANAGEMENT 2020. [DOI: 10.2217/bmt-2020-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Current targeted therapies for breast cancer include hormone inhibitors, monoclonal antibodies and tyrosine kinase inhibitors. However, a significant unmet therapeutic need remains for refractory disease and in particular for the triple negative subtype, which lacks hormone receptors and HER2. Chimeric antigen receptors T cells are genetically engineered to deploy selective cytolytic activity against cells that express cognate native target. Durable remissions have been achieved in refractory hematological malignancies but similar success against solid tumors remains elusive. Several hurdles hinder progress, including the need to identify safe antigens, promote T-cell homing to tumor sites and to ensure the persistence of functional chimeric antigen receptors T cells within the immunosuppressive tumor microenvironment. Perspectives to enable the attainment of this goal are presented in this review.
Collapse
Affiliation(s)
- Pierre Antoine
- King's College London, School of Cancer & Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London, SE1 9RT, UK
| | - John Maher
- King's College London, School of Cancer & Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London, SE1 9RT, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London, SE1 9RT, UK
- Department of Clinical Immunology & Allergy, King’s College Hospital NHS Foundation Trust, Denmark Hill, London, SE5 9RS, UK
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex, BN21 2UD, UK
| |
Collapse
|
148
|
Salzer B, Schueller CM, Zajc CU, Peters T, Schoeber MA, Kovacic B, Buri MC, Lobner E, Dushek O, Huppa JB, Obinger C, Putz EM, Holter W, Traxlmayr MW, Lehner M. Engineering AvidCARs for combinatorial antigen recognition and reversible control of CAR function. Nat Commun 2020; 11:4166. [PMID: 32820173 PMCID: PMC7441178 DOI: 10.1038/s41467-020-17970-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
T cells engineered to express chimeric antigen receptors (CAR-T cells) have shown impressive clinical efficacy in the treatment of B cell malignancies. However, the development of CAR-T cell therapies for solid tumors is hampered by the lack of truly tumor-specific antigens and poor control over T cell activity. Here we present an avidity-controlled CAR (AvidCAR) platform with inducible and logic control functions. The key is the combination of (i) an improved CAR design which enables controlled CAR dimerization and (ii) a significant reduction of antigen-binding affinities to introduce dependence on bivalent interaction, i.e. avidity. The potential and versatility of the AvidCAR platform is exemplified by designing ON-switch CARs, which can be regulated with a clinically applied drug, and AND-gate CARs specifically recognizing combinations of two antigens. Thus, we expect that AvidCARs will be a highly valuable platform for the development of controllable CAR therapies with improved tumor specificity.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Cytotoxicity, Immunologic/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/immunology
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Benjamin Salzer
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria
| | | | - Charlotte U Zajc
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria
| | - Timo Peters
- Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael A Schoeber
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Boris Kovacic
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Michelle C Buri
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Elisabeth Lobner
- Department of Biotechnology, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Omer Dushek
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Johannes B Huppa
- Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Christian Obinger
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Eva M Putz
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
| | - Wolfgang Holter
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria
- Department of Pediatrics, St. Anna Kinderspital, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael W Traxlmayr
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria.
- Department of Chemistry, Institute of Biochemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria.
| | - Manfred Lehner
- St. Anna Children's Cancer Research Institute (CCRI), 1090, Vienna, Austria.
- Christian Doppler Laboratory for Next Generation CAR T Cells, 1090, Vienna, Austria.
- Department of Pediatrics, St. Anna Kinderspital, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
149
|
Gavriil A, Barisa M, Halliwell E, Anderson J. Engineering Solutions for Mitigation of Chimeric Antigen Receptor T-Cell Dysfunction. Cancers (Basel) 2020; 12:E2326. [PMID: 32824734 PMCID: PMC7463974 DOI: 10.3390/cancers12082326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/06/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
The clinical successes of chimeric antigen receptor (CAR)-T-cell therapy targeting cell surface antigens in B cell leukaemias and lymphomas has demonstrated the proof of concept that appropriately engineered T-cells have the capacity to destroy advanced cancer with long term remissions ensuing. Nevertheless, it has been significantly more problematic to effect long term clinical benefit in a solid tumour context. A major contributing factor to the clinical failure of CAR-T-cells in solid tumours has been named, almost interchangeably, as T-cell "dysfunction" or "exhaustion". While unhelpful ambiguity surrounds the term "dysfunction", "exhaustion" is canonically regarded as a pejorative term for T-cells. Recent understanding of T-cell developmental biology now identifies exhausted cells as vital for effective immune responses in the context of ongoing antigenic challenge. The purpose of this review is to explore the critical stages in the CAR-T-cell life-cycle and their various contributions to T-cell exhaustion. Through an appreciation of the predominant mechanisms of CAR-T-cell exhaustion and resultant dysfunction, we describe a range of engineering approaches to improve CAR-T-cell function.
Collapse
Affiliation(s)
| | | | | | - John Anderson
- UCL Great Ormond Street, Institute of Child Health, London WC1N 1EH, UK; (A.G.); (M.B.); (E.H.)
| |
Collapse
|
150
|
Venetis K, Invernizzi M, Sajjadi E, Curigliano G, Fusco N. Cellular immunotherapy in breast cancer: The quest for consistent biomarkers. Cancer Treat Rev 2020; 90:102089. [PMID: 32889360 DOI: 10.1016/j.ctrv.2020.102089] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most common malignancy in women worldwide, with a relatively high proportion of patients experiencing resistance to standard treatments. Cellular immunotherapy (CI), which is based on the extraction, modification, and re-infusion of the patient's immune cells, is showing promising results in these patients. Among CI possible approaches, adoptive cell therapy (ACT) and dendritic cell (DC) vaccination are the most comprehensively explored in both primary/translational research studies and clinical trials. ACT may include the use of tumor-infiltrating lymphocytes (TILs), T cell receptor (TCR)-, or chimeric antigen receptor (CAR)-engineered T-cells. There are indications suggesting that a biomarker-based approach might be beneficial in effectively selecting breast cancer patients for CI. Here, we sought to provide the current knowledge of CI in breast cancer, focusing on candidate biomarkers, ongoing clinical trials, limitations, and immediate future perspectives.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Ph.D. Program in Translational Medicine, University of Milan, 20133 Milan, Italy; Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Marco Invernizzi
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Elham Sajjadi
- Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy.
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy.
| |
Collapse
|