51
|
Building a CAR-Treg: Going from the basic to the luxury model. Cell Immunol 2020; 358:104220. [DOI: 10.1016/j.cellimm.2020.104220] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 01/10/2023]
|
52
|
Radichev IA, Yoon J, Scott DW, Griffin K, Savinov AY. Towards antigen-specific Tregs for type 1 diabetes: Construction and functional assessment of pancreatic endocrine marker, HPi2-based chimeric antigen receptor. Cell Immunol 2020; 358:104224. [PMID: 33068914 DOI: 10.1016/j.cellimm.2020.104224] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/05/2023]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease marked by direct elimination of insulin-producing β cells by autoreactive T effectors. Recent T1D clinical trials utilizing autologous Tregs transfers to restore immune balance and improve disease has prompted us to design a novel Tregs-based antigen-specific T1D immunotherapy. We engineered a Chimeric Antigen Receptor (CAR) expressing a single-chain Fv recognizing the human pancreatic endocrine marker, HPi2. Human T cells, transduced with the resultant HPi2-CAR, proliferated and amplified Granzyme B accumulation when co-cultured with human, but not mouse β cells. Furthermore, following exposure of HPi2-CAR transduced cells to islets, CD8+ lymphocytes demonstrated enhanced CD107a (LAMP-1) expression, while CD4+ cells produced increased levels of IL-2. HPi2-CAR Tregs failed to maintain expansion due to a persistent tonic signaling from the CAR engagement to unexpectantly HPi2 antigen present on Tregs. Overall, we show lack of functionality of HPi2-CAR and highlight the importance of careful selection of CAR recognition driver for the sustainable activity and expandability of engineered T cells.
Collapse
Affiliation(s)
- Ilian A Radichev
- Diabetes Research Group/Sanford Research, Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD, USA
| | - Jeongheon Yoon
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - David W Scott
- Department of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Kurt Griffin
- Diabetes Research Group/Sanford Research, Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD, USA
| | - Alexei Y Savinov
- Diabetes Research Group/Sanford Research, Department of Pediatrics, University of South Dakota School of Medicine, Sioux Falls, SD, USA.
| |
Collapse
|
53
|
Rana J, Biswas M. Regulatory T cell therapy: Current and future design perspectives. Cell Immunol 2020; 356:104193. [PMID: 32823038 DOI: 10.1016/j.cellimm.2020.104193] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Regulatory T cells (Tregs) maintain immune equilibrium by suppressing immune responses through various multistep contact dependent and independent mechanisms. Cellular therapy using polyclonal Tregs in transplantation and autoimmune diseases has shown promise in preclinical models and clinical trials. Although novel approaches have been developed to improve specificity and efficacy of antigen specific Treg based therapies, widespread application is currently restricted. To date, design-based approaches to improve the potency and persistence of engineered chimeric antigen receptor (CAR) Tregs are limited. Here, we describe currently available Treg based therapies, their advantages and limitations for implementation in clinical studies. We also examine various strategies for improving CAR T cell design that can potentially be applied to CAR Tregs, such as identifying co-stimulatory signalling domains that enhance suppressive ability, determining optimal scFv affinity/avidity, and co-expression of accessory molecules. Finally, we discuss the importance of tailoring CAR Treg design to suit the individual disease.
Collapse
Affiliation(s)
- Jyoti Rana
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
54
|
Zhang PF, Xie D, Li Q. Chimeric antigen receptor T-cell therapy beyond cancer: current practice and future prospects. Immunotherapy 2020; 12:1021-1034. [PMID: 32727249 DOI: 10.2217/imt-2020-0009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor T (CAR-T) cells has achieved remarkable efficacy in the treatment of hematological malignancies, which has inspired researchers to expand the application of CAR-T-cell therapy to other medical conditions. Here, we review the current understanding and development of CAR-T-cell therapy for infectious diseases, autoimmune diseases and allotransplantation. The limitations and challenges of CAR-T-cell therapy in the treatment of these diseases and potential solutions to overcome these shortcomings are also discussed. With the development of novel designs of CARs and preclinical/clinical investigations, CAR-T-cell therapy is expected to be a potential cure option in a wide array of disease settings in the future.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China, 610041
| | - Dan Xie
- Prenatal Diagnosis Center, Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China, 610041.,Key Laboratory of Birth Defects & Related Diseases of Women & Children (Sichuan University), Ministry of Education, Chengdu, China, 610041
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China, 610041
| |
Collapse
|
55
|
Mohseni YR, Tung SL, Dudreuilh C, Lechler RI, Fruhwirth GO, Lombardi G. The Future of Regulatory T Cell Therapy: Promises and Challenges of Implementing CAR Technology. Front Immunol 2020; 11:1608. [PMID: 32793236 PMCID: PMC7393941 DOI: 10.3389/fimmu.2020.01608] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022] Open
Abstract
Cell therapy with polyclonal regulatory T cells (Tregs) has been translated into the clinic and is currently being tested in transplant recipients and patients suffering from autoimmune diseases. Moreover, building on animal models, it has been widely reported that antigen-specific Tregs are functionally superior to polyclonal Tregs. Among various options to confer target specificity to Tregs, genetic engineering is a particularly timely one as has been demonstrated in the treatment of hematological malignancies where it is in routine clinical use. Genetic engineering can be exploited to express chimeric antigen receptors (CAR) in Tregs, and this has been successfully demonstrated to be robust in preclinical studies across various animal disease models. However, there are several caveats and a number of strategies should be considered to further improve on targeting, efficacy and to understand the in vivo distribution and fate of CAR-Tregs. Here, we review the differing approaches to confer antigen specificity to Tregs with emphasis on CAR-Tregs. This includes an overview and discussion of the various approaches to improve CAR-Treg specificity and therapeutic efficacy as well as addressing potential safety concerns. We also discuss different imaging approaches to understand the in vivo biodistribution of administered Tregs. Preclinical research as well as suitability of methodologies for clinical translation are discussed.
Collapse
MESH Headings
- Animals
- Antigens/immunology
- Bioengineering
- Humans
- Immunomodulation
- Immunotherapy, Adoptive/methods
- 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-Cell Antigen Receptor Specificity
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Yasmin R. Mohseni
- Peter Gorer Department of Immunobiology, MRC Centre for Transplantation, School of Immunology and Microbial Science, King's College London (KCL), Guy's Hospital, London, United Kingdom
| | - Sim L. Tung
- Peter Gorer Department of Immunobiology, MRC Centre for Transplantation, School of Immunology and Microbial Science, King's College London (KCL), Guy's Hospital, London, United Kingdom
| | - Caroline Dudreuilh
- Peter Gorer Department of Immunobiology, MRC Centre for Transplantation, School of Immunology and Microbial Science, King's College London (KCL), Guy's Hospital, London, United Kingdom
| | - Robert I. Lechler
- Peter Gorer Department of Immunobiology, MRC Centre for Transplantation, School of Immunology and Microbial Science, King's College London (KCL), Guy's Hospital, London, United Kingdom
| | - Gilbert O. Fruhwirth
- Imaging Therapies and Cancer Group, Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, MRC Centre for Transplantation, School of Immunology and Microbial Science, King's College London (KCL), Guy's Hospital, London, United Kingdom
| |
Collapse
|
56
|
Carballido JM, Regairaz C, Rauld C, Raad L, Picard D, Kammüller M. The Emerging Jamboree of Transformative Therapies for Autoimmune Diseases. Front Immunol 2020; 11:472. [PMID: 32296421 PMCID: PMC7137386 DOI: 10.3389/fimmu.2020.00472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
Standard treatments for autoimmune and autoinflammatory disorders rely mainly on immunosuppression. These are predominantly symptomatic remedies that do not affect the root cause of the disease and are associated with multiple side effects. Immunotherapies are being developed during the last decades as more specific and safer alternatives to small molecules with broad immunosuppressive activity, but they still do not distinguish between disease-causing and protective cell targets and thus, they still have considerable risks of increasing susceptibility to infections and/or malignancy. Antigen-specific approaches inducing immune tolerance represent an emerging trend carrying the potential to be curative without inducing broad immunosuppression. These therapies are based on antigenic epitopes derived from the same proteins that are targeted by the autoreactive T and B cells, and which are administered to patients together with precise instructions to induce regulatory responses capable to restore homeostasis. They are not personalized medicines, and they do not need to be. They are precision therapies exquisitely targeting the disease-causing cells that drive pathology in defined patient populations. Immune tolerance approaches are truly transformative options for people suffering from autoimmune diseases.
Collapse
Affiliation(s)
- José M. Carballido
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Camille Regairaz
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Celine Rauld
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Layla Raad
- Autoimmunity Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Damien Picard
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael Kammüller
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
57
|
El-Ayachi I, Washburn WK, Schenk AD. Recent Progress in Treg Biology and Transplant Therapeutics. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00278-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Abstract
Purpose of Review
Regulatory T cell (Treg) biology continues to evolve at a rapid pace. The role of Tregs in solid organ transplantation offers a unique window into Treg ontogeny and function as well as limitless possibilities for clinical application. Here we review recent significant discoveries and key translational work.
Recent Findings
Advances in transplantation deepen understanding of Treg differentiation, expansion, transcription, co-stimulation, and signaling. T cell receptor (TCR) sequencing and single-cell analytics allow unprecedented insight into Treg repertoire diversity and phenotypic heterogeneity. Efforts to replace conventional immunosuppression with Treg adoptive immunotherapy are underway and coalescing around strategies to increase efficiency through development of donor-reactive Tregs.
Summary
Adoptive immunotherapy with Tregs is a leading tolerogenic strategy. Early clinical trials suggest that Treg infusion is safe and reports on efficacy will soon follow.
Collapse
|
58
|
Boardman DA, Levings MK. Cancer immunotherapies repurposed for use in autoimmunity. Nat Biomed Eng 2019; 3:259-263. [PMID: 30952977 DOI: 10.1038/s41551-019-0359-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Dominic A Boardman
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada. .,BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada. .,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
59
|
Mancusi A, Piccinelli S, Velardi A, Pierini A. CD4 +FOXP3 + Regulatory T Cell Therapies in HLA Haploidentical Hematopoietic Transplantation. Front Immunol 2019; 10:2901. [PMID: 31921162 PMCID: PMC6927932 DOI: 10.3389/fimmu.2019.02901] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022] Open
Abstract
Since their discovery CD4+FOXP3+ regulatory T cells (Tregs) represented a promising tool to induce tolerance in allogeneic hematopoietic cell transplantation. Preclinical models proved that adoptive transfer of Tregs or the use of compounds that can favor their function in vivo are effective for prevention and treatment of graft-vs.-host disease (GvHD). Following these findings, Treg-based therapies have been employed in clinical trials. Adoptive immunotherapy with Tregs effectively prevents GvHD induced by alloreactive T cells in the setting of one HLA haplotype mismatched hematopoietic transplantation. The absence of post transplant pharmacologic immunosuppression unleashes T-cell mediated graft-vs.-tumor (GvT) effect, which results in an unprecedented, almost complete control of leukemia relapse in this setting. In the present review, we will report preclinical studies and clinical trials that demonstrate Treg ability to promote donor engraftment, protect from GvHD and improve GvT effect. We will also discuss new strategies to further enhance in vivo efficacy of Treg-based therapies.
Collapse
Affiliation(s)
- Antonella Mancusi
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| | - Sara Piccinelli
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| | - Antonio Pierini
- Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
60
|
Yu S, Su C, Luo X. Impact of infection on transplantation tolerance. Immunol Rev 2019; 292:243-263. [PMID: 31538351 PMCID: PMC6961566 DOI: 10.1111/imr.12803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
Allograft tolerance is the ultimate goal of organ transplantation. Current strategies for tolerance induction mainly focus on inhibiting alloreactive T cells while promoting regulatory immune cells. Pathogenic infections may have direct impact on both effector and regulatory cell populations, therefore can alter host susceptibility to transplantation tolerance induction as well as impair the quality and stability of tolerance once induced. In this review, we will discuss existing data demonstrating the effect of infections on transplantation tolerance, with particular emphasis on the role of the stage of infection (acute, chronic, or latent) and the stage of tolerance (induction or maintenance) in this infection-tolerance interaction. While the deleterious effect of acute infection on tolerance is mainly driven by proinflammatory cytokines induced shortly after the infection, chronic infection may generate exhausted T cells that could in fact facilitate transplantation tolerance. In addition to pathogenic infections, commensal intestinal microbiota also has numerous significant immunomodulatory effects that can shape the host alloimmunity following transplantation. A comprehensive understanding of these mechanisms is crucial for the development of therapeutic strategies for robustly inducing and stably maintaining transplantation tolerance while preserving host anti-pathogen immunity in clinically relevant scenarios.
Collapse
Affiliation(s)
- Shuangjin Yu
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Division of Organ transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chang Su
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
61
|
Flippe L, Bézie S, Anegon I, Guillonneau C. Future prospects for CD8 + regulatory T cells in immune tolerance. Immunol Rev 2019; 292:209-224. [PMID: 31593314 PMCID: PMC7027528 DOI: 10.1111/imr.12812] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD8+ Tregs have been long described and significant progresses have been made about their phenotype, their functional mechanisms, and their suppressive ability compared to conventional CD4+ Tregs. They are now at the dawn of their clinical use. In this review, we will summarize their phenotypic characteristics, their mechanisms of action, the similarities, differences and synergies between CD8+ and CD4+ Tregs, and we will discuss the biology, development and induction of CD8+ Tregs, their manufacturing for clinical use, considering open questions/uncertainties and future technically accessible improvements notably through genetic modifications.
Collapse
Affiliation(s)
- Léa Flippe
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
62
|
Cabello-Olmo M, Araña M, Radichev I, Smith P, Huarte E, Barajas M. New Insights into Immunotherapy Strategies for Treating Autoimmune Diabetes. Int J Mol Sci 2019; 20:ijms20194789. [PMID: 31561568 PMCID: PMC6801436 DOI: 10.3390/ijms20194789] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/18/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes mellitus (T1D) is an autoimmune illness that affects millions of patients worldwide. The main characteristic of this disease is the destruction of pancreatic insulin-producing beta cells that occurs due to the aberrant activation of different immune effector cells. Currently, T1D is treated by lifelong administration of novel versions of insulin that have been developed recently; however, new approaches that could address the underlying mechanisms responsible for beta cell destruction have been extensively investigated. The strategies based on immunotherapies have recently been incorporated into a panel of existing treatments for T1D, in order to block T-cell responses against beta cell antigens that are very common during the onset and development of T1D. However, a complete preservation of beta cell mass as well as insulin independency is still elusive. As a result, there is no existing T1D targeted immunotherapy able to replace standard insulin administration. Presently, a number of novel therapy strategies are pursuing the goals of beta cell protection and normoglycemia. In the present review we explore the current state of immunotherapy in T1D by highlighting the most important studies in this field, and envision novel strategies that could be used to treat T1D in the future.
Collapse
Affiliation(s)
- Miriam Cabello-Olmo
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, 31008 Pamplona, Spain.
| | - Miriam Araña
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, 31008 Pamplona, Spain.
| | - Ilian Radichev
- Diabetes research group at Sanford Research, Sioux Falls, SD 57104, USA.
| | - Paul Smith
- Incyte Corporation, Wilmington, DE 19803, USA.
| | | | - Miguel Barajas
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
63
|
Abstract
Chimeric antigen receptors (CARs) have shown remarkable ability to re-direct T cells to target CD19-expressing tumours, resulting in remission rates of up to 90% in individuals with paediatric acute lymphoblastic lymphoma. Lessons learned from these clinical trials of adoptive T cell therapy for cancer, as well as investments made in manufacturing T cells at commercial scale, have inspired researchers to develop CARs for additional applications. Here, we explore the challenges and opportunities of using this technology to target infectious diseases such as with HIV and undesired immune responses such as autoimmunity and transplant rejection. Despite substantial obstacles, the potential of CAR T cells to enable cures for a wide array of disease settings could be transformational for the medical field.
Collapse
|
64
|
Pierini A, Ruggeri L, Mancusi A, Carotti A, Falzetti F, Terenzi A, Martelli MF, Velardi A. T cell depletion and no post transplant immune suppression allow separation of graft versus leukemia from graft versus host disease. Bone Marrow Transplant 2019; 54:775-779. [DOI: 10.1038/s41409-019-0597-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
65
|
Meyer EH, Laport G, Xie BJ, MacDonald K, Heydari K, Sahaf B, Tang SW, Baker J, Armstrong R, Tate K, Tadisco C, Arai S, Johnston L, Lowsky R, Muffly L, Rezvani AR, Shizuru J, Weng WK, Sheehan K, Miklos D, Negrin RS. Transplantation of donor grafts with defined ratio of conventional and regulatory T cells in HLA-matched recipients. JCI Insight 2019; 4:127244. [PMID: 31092732 DOI: 10.1172/jci.insight.127244] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDIn preclinical murine and early clinical studies of hematopoietic cell transplantation, engineering of donor grafts with defined ratios of CD4+CD25+FoxP3+ Tregs to conventional T cells (Tcons) results in the prevention of graft-versus-host disease and improved immune reconstitution. The use of highly purified primary graft Tregs for direct cell infusion has potential advantages over impure immunomagnetic selection or culture expansion, but has not been tested clinically. We performed a phase I study of the timed addition of CD34-selected hematopoietic stem cells and Tregs, followed by Tcons for the treatment of patients with high-risk hematological malignancies.METHODSWe present interim evaluation of a single-center open phase I/II study of administration of human leukocyte-matched Tregs and CD34-selected hematopoietic cells, followed by infusion of an equal ratio of Tcons in adult patients undergoing myeloablative hematopoietic stem cell transplantation (HCT) for high-risk or active hematological malignancies. Tregs were purified by immunomagnetic selection and high-speed cell sorting.RESULTSHere we report results for the first 12 patients who received Tregs of between 91% and 96% purity. Greater than grade II GVHD was noted in 2 patients in the first cohort of 5 patients, who received cryopreserved Tregs, but neither acute nor chronic GVHD was noted in the second cohort of 7 patients, who received fresh Tregs and single-agent GVHD prophylaxis. Patients in the second cohort appeared to have normal immune reconstitution compared with patients who underwent transplantation and did not develop GVHD.CONCLUSIONOur study shows that the use of highly purified fresh Tregs is clinically feasible and supports continued investigation of the strategy.TRIAL REGISTRATIONClinicalTrials.gov NCT01660607.FUNDINGNIH NHBLI R01 HL114591 and K08HL119590.
Collapse
Affiliation(s)
- Everett H Meyer
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA.,Cell Therapy Facility, Stanford Health Care, Stanford, California, USA
| | - Ginna Laport
- Tempest Therapeutics, San Francisco, California, USA
| | - Bryan J Xie
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Kate MacDonald
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Kartoosh Heydari
- Cell Therapy Facility, Stanford Health Care, Stanford, California, USA
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Sai-Wen Tang
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Randall Armstrong
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Keri Tate
- Laboratory for Cell and Gene Medicine, Stanford University, Palo Alto, California, USA
| | - Cynthia Tadisco
- Laboratory for Cell and Gene Medicine, Stanford University, Palo Alto, California, USA
| | - Sally Arai
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Laura Johnston
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Robert Lowsky
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Lori Muffly
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Andrew R Rezvani
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Kevin Sheehan
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - David Miklos
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, California, USA
| |
Collapse
|
66
|
Dawson NA, Lamarche C, Hoeppli RE, Bergqvist P, Fung VC, McIver E, Huang Q, Gillies J, Speck M, Orban PC, Bush JW, Mojibian M, Levings MK. Systematic testing and specificity mapping of alloantigen-specific chimeric antigen receptors in regulatory T cells. JCI Insight 2019; 4:123672. [PMID: 30753169 DOI: 10.1172/jci.insight.123672] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/05/2019] [Indexed: 12/19/2022] Open
Abstract
Chimeric antigen receptor (CAR) technology can be used to engineer the antigen specificity of regulatory T cells (Tregs) and improve their potency as an adoptive cell therapy in multiple disease models. As synthetic receptors, CARs carry the risk of immunogenicity, particularly when derived from nonhuman antibodies. Using an HLA-A*02:01-specific CAR (A2-CAR) encoding a single-chain variable fragment (Fv) derived from a mouse antibody, we developed a panel of 20 humanized A2-CARs (hA2-CARs). Systematic testing demonstrated variations in expression, and ability to bind HLA-A*02:01 and stimulate human Treg suppression in vitro. In addition, we developed a new method to comprehensively map the alloantigen specificity of CARs, revealing that humanization reduced HLA-A cross-reactivity. In vivo bioluminescence imaging showed rapid trafficking and persistence of hA2-CAR Tregs in A2-expressing allografts, with eventual migration to draining lymph nodes. Adoptive transfer of hA2-CAR Tregs suppressed HLA-A2+ cell-mediated xenogeneic graft-versus-host disease and diminished rejection of human HLA-A2+ skin allografts. These data provide a platform for systematic development and specificity testing of humanized alloantigen-specific CARs that can be used to engineer specificity and homing of therapeutic Tregs.
Collapse
Affiliation(s)
- Nicholas Aj Dawson
- Department of Medicine and.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Caroline Lamarche
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Romy E Hoeppli
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Peter Bergqvist
- Centre for Drug and Research and Development, Vancouver, British Columbia, Canada
| | - Vivian Cw Fung
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Emma McIver
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Qing Huang
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Jana Gillies
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Madeleine Speck
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Paul C Orban
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Jonathan W Bush
- BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine and
| | - Majid Mojibian
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia (UBC), Vancouver, British Columbia, Canada.,BC Children's Hospital Research Institute (BCCHR), Vancouver, British Columbia, Canada.,School of Biomedical Engineering, UBC, Vancouver, British Columbia, Canada
| |
Collapse
|
67
|
Abstract
Breakthroughs in gene synthesis has allowed synthetic biologists the ability to design any DNA sequence of interest, enabling the possibility to create complex systems inside cells with novel functions to tackle problems in immunology. Synthetic immunology of mammalian cells expressing natural or synthetic genes can guide and induce immune responses in patients. Through recent developments in engineering chimeric receptors, it is now feasible to customize control over engineered cells to target the disease sites with specificity. These cells can avoid immune rejection if derived from expandable cell types (e.g., stem cells or T cells) and then can be grown in abundance before implantation. However, safety concerns of engineered cells in circulation necessitates the development of a wide range of mechanisms to kill cells after their therapeutic life ends. This therapeutic effect is still predominantly the secretion of therapeutic proteins, but novel therapeutic interventions have been explored by synthetic biologists. In the pursuit of engineering new cell functions for synthetic immunology, it is possible that many problems previously thought intractable may actually be possible.
Collapse
Affiliation(s)
- Niema Binth Mohammad
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Candice Chee Ka Lam
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Kevin Truong
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada.,Edward S. Rogers, Sr. Department of Electrical and Computer Engineering , University of Toronto , 10 King's College Circle , Toronto , Ontario M5S 3G4 , Canada
| |
Collapse
|
68
|
Gupta PK, McIntosh CM, Chong AS, Alegre ML. The pursuit of transplantation tolerance: new mechanistic insights. Cell Mol Immunol 2019; 16:324-333. [PMID: 30760917 DOI: 10.1038/s41423-019-0203-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/17/2019] [Indexed: 12/11/2022] Open
Abstract
Donor-specific transplantation tolerance that enables weaning from immunosuppressive drugs but retains immune competence to non-graft antigens has been a lasting pursuit since the discovery of neonatal tolerance. More recently, efforts have been devoted not only to understanding how transplantation tolerance can be induced but also the mechanisms necessary to maintain it as well as how inflammatory exposure challenges its durability. This review focuses on recent advances regarding key peripheral mechanisms of T cell tolerance, with the underlying hypothesis that a combination of several of these mechanisms may afford a more robust and durable tolerance and that a better understanding of these individual pathways may permit longitudinal tracking of tolerance following clinical transplantation to serve as biomarkers. This review may enable a personalized assessment of the degree of tolerance in individual patients and the opportunity to strengthen the robustness of peripheral tolerance.
Collapse
Affiliation(s)
- Pawan K Gupta
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | | | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, 60637, USA
| | - Maria-Luisa Alegre
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
69
|
Zhang Q, Lu W, Liang CL, Chen Y, Liu H, Qiu F, Dai Z. Chimeric Antigen Receptor (CAR) Treg: A Promising Approach to Inducing Immunological Tolerance. Front Immunol 2018; 9:2359. [PMID: 30369931 PMCID: PMC6194362 DOI: 10.3389/fimmu.2018.02359] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/24/2018] [Indexed: 12/14/2022] Open
Abstract
Cellular therapies with polyclonal regulatory T-cells (Tregs) in transplantation and autoimmune diseases have been carried out in both animal models and clinical trials. However, The use of large numbers of polyclonal Tregs with unknown antigen specificities has led to unwanted effects, such as systemic immunosuppression, which can be avoided via utilization of antigen-specific Tregs. Antigen-specific Tregs are also more potent in suppression than polyclonal ones. Although antigen-specific Tregs can be induced in vitro, these iTregs are usually contaminated with effector T cells during in vitro expansion. Fortunately, Tregs can be efficiently engineered with a predetermined antigen-specificity via transfection of viral vectors encoding specific T cell receptors (TCRs) or chimeric antigen receptors (CARs). Compared to Tregs engineered with TCRs (TCR-Tregs), CAR-modified Tregs (CAR-Tregs) engineered in a non-MHC restricted manner have the advantage of widespread applications, especially in transplantation and autoimmunity. CAR-Tregs also are less dependent on IL-2 than are TCR-Tregs. CAR-Tregs are promising given that they maintain stable phenotypes and functions, preferentially migrate to target sites, and exert more potent and specific immunosuppression than do polyclonal Tregs. However, there are some major hurdles that must be overcome before CAR-Tregs can be used in clinic. It is known that treatments with anti-tumor CAR-T cells cause side effects due to cytokine “storm” and neuronal cytotoxicity. It is unclear whether CAR-Tregs would also induce these adverse reactions. Moreover, antibodies specific for self- or allo-antigens must be characterized to construct antigen-specific CAR-Tregs. Selection of antigens targeted by CARs and development of specific antibodies are difficult in some disease models. Finally, CAR-Treg exhaustion may limit their efficacy in immunosuppression. Recently, innovative CAR-Treg therapies in animal models of transplantation and autoimmune diseases have been reported. In this mini-review, we have summarized recent progress of CAR-Tregs and discussed their potential applications for induction of immunological tolerance.
Collapse
Affiliation(s)
- Qunfang Zhang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Weihui Lu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chun-Ling Liang
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuchao Chen
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Huazhen Liu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Feifei Qiu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhenhua Dai
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences, and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
70
|
Biswas M, Kumar SRP, Terhorst C, Herzog RW. Gene Therapy With Regulatory T Cells: A Beneficial Alliance. Front Immunol 2018; 9:554. [PMID: 29616042 PMCID: PMC5868074 DOI: 10.3389/fimmu.2018.00554] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 03/05/2018] [Indexed: 12/12/2022] Open
Abstract
Gene therapy aims to replace a defective or a deficient protein at therapeutic or curative levels. Improved vector designs have enhanced safety, efficacy, and delivery, with potential for lasting treatment. However, innate and adaptive immune responses to the viral vector and transgene product remain obstacles to the establishment of therapeutic efficacy. It is widely accepted that endogenous regulatory T cells (Tregs) are critical for tolerance induction to the transgene product and in some cases the viral vector. There are two basic strategies to harness the suppressive ability of Tregs: in vivo induction of adaptive Tregs specific to the introduced gene product and concurrent administration of autologous, ex vivo expanded Tregs. The latter may be polyclonal or engineered to direct specificity to the therapeutic antigen. Recent clinical trials have advanced adoptive immunotherapy with Tregs for the treatment of autoimmune disease and in patients receiving cell transplants. Here, we highlight the potential benefit of combining gene therapy with Treg adoptive transfer to achieve a sustained transgene expression. Furthermore, techniques to engineer antigen-specific Treg cell populations, either through reprogramming conventional CD4+ T cells or transferring T cell receptors with known specificity into polyclonal Tregs, are promising in preclinical studies. Thus, based upon these observations and the successful use of chimeric (IgG-based) antigen receptors (CARs) in antigen-specific effector T cells, different types of CAR-Tregs could be added to the repertoire of inhibitory modalities to suppress immune responses to therapeutic cargos of gene therapy vectors. The diverse approaches to harness the ability of Tregs to suppress unwanted immune responses to gene therapy and their perspectives are reviewed in this article.
Collapse
Affiliation(s)
- Moanaro Biswas
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Sandeep R P Kumar
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, United States
| | - Roland W Herzog
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
71
|
Marshall GP, Cserny J, Perry DJ, Yeh WI, Seay HR, Elsayed AG, Posgai AL, Brusko TM. Clinical Applications of Regulatory T cells in Adoptive Cell Therapies. CELL & GENE THERAPY INSIGHTS 2018; 4:405-429. [PMID: 34984106 PMCID: PMC8722436 DOI: 10.18609/cgti.2018.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Interest in adoptive T-cell therapies has been ignited by the recent clinical success of genetically-modified T cells in the cancer immunotherapy space. In addition to immune targeting for malignancies, this approach is now being explored for the establishment of immune tolerance with regulatory T cells (Tregs). Herein, we will summarize the basic science and clinical results emanating from trials directed at inducing durable immune regulation through administration of Tregs. We will discuss some of the current challenges facing the field in terms of maximizing cell purity, stability and expansion capacity, while also achieving feasibility and GMP production. Indeed, recent advances in methodologies for Treg isolation, expansion, and optimal source materials represent important strides toward these considerations. Finally, we will review the emerging genetic and biomaterial-based approaches on the horizon for directing Treg specificity to augment tissue-targeting and regenerative medicine.
Collapse
Affiliation(s)
| | - Judit Cserny
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Daniel J Perry
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Wen-I Yeh
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Howard R Seay
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Ahmed G Elsayed
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA.,Department of Microbiology and Immunology, Faculty of Medicine, Mansoura University, Egypt
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Todd M Brusko
- OneVax LLC, Sid Martin Biotechnology Institute, Alachua, Florida, USA.,Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|