1
|
Zhong Y, Stauss HJ. Targeted Therapy of Multiple Sclerosis: A Case for Antigen-Specific Tregs. Cells 2024; 13:797. [PMID: 38786021 PMCID: PMC11119434 DOI: 10.3390/cells13100797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
Multiple sclerosis is an autoinflammatory condition that results in damage to myelinated neurons in affected patients. While disease-modifying treatments have been successful in slowing the progression of relapsing-remitting disease, most patients still progress to secondary progressive disease that is largely unresponsive to disease-modifying treatments. Similarly, there is currently no effective treatment for patients with primary progressive MS. Innate and adaptive immune cells in the CNS play a critical role in initiating an autoimmune attack and in maintaining the chronic inflammation that drives disease progression. In this review, we will focus on recent insights into the role of T cells with regulatory function in suppressing the progression of MS, and, more importantly, in promoting the remyelination and repair of MS lesions in the CNS. We will discuss the exciting potential to genetically reprogram regulatory T cells to achieve immune suppression and enhance repair locally at sites of tissue damage, while retaining a fully competent immune system outside the CNS. In the future, reprogramed regulatory T cells with defined specificity and function may provide life medicines that can persist in patients and achieve lasting disease suppression after one cycle of treatment.
Collapse
Affiliation(s)
| | - Hans J. Stauss
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PP, UK;
| |
Collapse
|
2
|
Azad A, Altunbas HA, Manguoglu AE. From islet transplantation to beta-cell regeneration: an update on beta-cell-based therapeutic approaches in type 1 diabetes. Expert Rev Endocrinol Metab 2024; 19:217-227. [PMID: 38693782 DOI: 10.1080/17446651.2024.2347263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 03/06/2024] [Indexed: 05/03/2024]
Abstract
INTRODUCTION Type 1 diabetes (T1D) mellitus is an autoimmune disease in which immune cells, predominantly effector T cells, destroy insulin-secreting beta-cells. Beta-cell destruction led to various consequences ranging from retinopathy and nephropathy to neuropathy. Different strategies have been developed to achieve normoglycemia, including exogenous glucose compensation, whole pancreas transplantation, islet transplantation, and beta-cell replacement. AREAS COVERED The last two decades of experience have shown that indigenous glucose compensation through beta-cell regeneration and protection is a peerless method for T1D therapy. Tremendous studies have tried to find an unlimited source for beta-cell regeneration, on the one hand, and beta-cell protection against immune attack, on the other hand. Recent advances in stem cell technology, gene editing methods, and immune modulation approaches provide a unique opportunity for both beta-cell regeneration and protection. EXPERT OPINION Pluripotent stem cell differentiation into the beta-cell is considered an unlimited source for beta-cell regeneration. Devising engineered pancreas-specific regulatory T cells using Chimeric Antigen Receptor (CAR) technology potentiates an effective immune tolerance induction for beta-cell protection. Beta-cell regeneration using pluripotent stem cells and beta-cell protection using pancreas-specific engineered regulatory T cells promises to develop a curative protocol in T1D.
Collapse
Affiliation(s)
- Asef Azad
- Department of Medical Biology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Hasan Ali Altunbas
- Department of Endocrinology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ayse Esra Manguoglu
- Department of Medical Biology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
3
|
Bittner S, Hehlgans T, Feuerer M. Engineered Treg cells as putative therapeutics against inflammatory diseases and beyond. Trends Immunol 2023; 44:468-483. [PMID: 37100644 DOI: 10.1016/j.it.2023.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/28/2023]
Abstract
Regulatory T (Treg) cells ensure tolerance against self-antigens, limit excessive inflammation, and support tissue repair processes. Therefore, Treg cells are currently attractive candidates for the treatment of certain inflammatory diseases, autoimmune disorders, or transplant rejection. Early clinical trials have proved the safety and efficacy of certain Treg cell therapies in inflammatory diseases. We summarize recent advances in engineering Treg cells, including the concept of biosensors for inflammation. We assess Treg cell engineering possibilities for novel functional units, including Treg cell modifications influencing stability, migration, and tissue adaptation. Finally, we outline perspectives of engineered Treg cells going beyond inflammatory diseases by using custom-designed receptors and read-out systems, aiming to use Treg cells as in vivo diagnostic tools and drug delivery vehicles.
Collapse
Affiliation(s)
- Sebastian Bittner
- Leibniz Institute for Immunotherapy, Division of Immunology, 93053 Regensburg, Germany
| | - Thomas Hehlgans
- Leibniz Institute for Immunotherapy, Division of Immunology, 93053 Regensburg, Germany; Chair for Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Markus Feuerer
- Leibniz Institute for Immunotherapy, Division of Immunology, 93053 Regensburg, Germany; Chair for Immunology, University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
4
|
Steiner R, Pilat N. The potential for Treg-enhancing therapies in transplantation. Clin Exp Immunol 2023; 211:122-137. [PMID: 36562079 PMCID: PMC10019131 DOI: 10.1093/cei/uxac118] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/21/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022] Open
Abstract
Since the discovery of regulatory T cells (Tregs) as crucial regulators of immune tolerance against self-antigens, these cells have become a promising tool for the induction of donor-specific tolerance in transplantation medicine. The therapeutic potential of increasing in vivoTreg numbers for a favorable Treg to Teff cell ratio has already been demonstrated in several sophisticated pre-clinical models and clinical pilot trials. In addition to improving cell quantity, enhancing Treg function utilizing engineering techniques led to encouraging results in models of autoimmunity and transplantation. Here we aim to discuss the most promising approaches for Treg-enhancing therapies, starting with adoptive transfer approaches and ex vivoexpansion cultures (polyclonal vs. antigen specific), followed by selective in vivostimulation methods. Furthermore, we address next generation concepts for Treg function enhancement (CARs, TRUCKs, BARs) as well as the advantages and caveats inherit to each approach. Finally, this review will discuss the clinical experience with Treg therapy in ongoing and already published clinical trials; however, data on long-term results and efficacy are still very limited and many questions that might complicate clinical translation remain open. Here, we discuss the hurdles for clinical translation and elaborate on current Treg-based therapeutic options as well as their potencies for improving long-term graft survival in transplantation.
Collapse
Affiliation(s)
- Romy Steiner
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Nina Pilat
- Correspondence: Nina Pilat, PhD, Department of Cardiac Surgery, Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
5
|
Yang SJ, Singh AK, Drow T, Tappen T, Honaker Y, Barahmand-Pour-Whitman F, Linsley PS, Cerosaletti K, Mauk K, Xiang Y, Smith J, Mortensen E, Cook PJ, Sommer K, Khan I, Liggitt D, Rawlings DJ, Buckner JH. Pancreatic islet-specific engineered T regs exhibit robust antigen-specific and bystander immune suppression in type 1 diabetes models. Sci Transl Med 2022; 14:eabn1716. [PMID: 36197963 DOI: 10.1126/scitranslmed.abn1716] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Adoptive transfer of regulatory T cells (Tregs) is therapeutic in type 1 diabetes (T1D) mouse models. Tregs that are specific for pancreatic islets are more potent than polyclonal Tregs in preventing disease. However, the frequency of antigen-specific natural Tregs is extremely low, and ex vivo expansion may destabilize Tregs, leading to an effector phenotype. Here, we generated durable, antigen-specific engineered Tregs (EngTregs) from primary human CD4+ T cells by combining FOXP3 homology-directed repair editing and lentiviral T cell receptor (TCR) delivery. Using TCRs derived from clonally expanded CD4+ T cells isolated from patients with T1D, we generated islet-specific EngTregs that suppressed effector T cell (Teff) proliferation and cytokine production. EngTregs suppressed Teffs recognizing the same islet antigen in addition to bystander Teffs recognizing other islet antigens through production of soluble mediators and both direct and indirect mechanisms. Adoptively transferred murine islet-specific EngTregs homed to the pancreas and blocked diabetes triggered by islet-specific Teffs or diabetogenic polyclonal Teffs in recipient mice. These data demonstrate the potential of antigen-specific EngTregs as a targeted therapy for preventing T1D.
Collapse
Affiliation(s)
- Soo Jung Yang
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Akhilesh K Singh
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Travis Drow
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Tori Tappen
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Yuchi Honaker
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Fariba Barahmand-Pour-Whitman
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Peter S Linsley
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Karen Cerosaletti
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Kelsey Mauk
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Yufei Xiang
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Jessica Smith
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Emma Mortensen
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Peter J Cook
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Karen Sommer
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Iram Khan
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA 98101, USA
| | - David J Rawlings
- Center for Immunity and Immunotherapies and the Program for Cell and Gene Therapy, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98101, USA.,Department of Immunology, University of Washington, Seattle, WA 98101, USA
| | - Jane H Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA.,Department of Immunology, University of Washington, Seattle, WA 98101, USA.,Department of Medicine, University of Washington, Seattle, WA 98101, USA
| |
Collapse
|
6
|
Yan S, Kotschenreuther K, Deng S, Kofler DM. Regulatory T cells in rheumatoid arthritis: functions, development, regulation, and therapeutic potential. Cell Mol Life Sci 2022; 79:533. [PMID: 36173485 PMCID: PMC9522664 DOI: 10.1007/s00018-022-04563-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/31/2022] [Accepted: 09/17/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that mainly affects the joints but also leads to systemic inflammation. Auto-reactivity and dysregulation of self-tolerance are thought to play a vital role in disease onset. In the pathogenesis of autoimmune diseases, disturbed immunosuppressive properties of regulatory T cells contribute to the dysregulation of immune homeostasis. In RA patients, the functions of Treg cells and their frequency are reduced. Therefore, focusing on the re-establishment of self-tolerance by increasing Treg cell frequencies and preventing a loss of function is a promising strategy for the treatment of RA. This approach could be especially beneficial for those patients who do not respond well to current therapies. In this review, we summarize and discuss the current knowledge about the function, differentiation and regulation of Treg cells in RA patients and in animal models of autoimmune arthritis. In addition, we highlight the therapeutic potential as well as the challenges of Treg cell targeting treatment strategies.
Collapse
Affiliation(s)
- Shuaifeng Yan
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Konstantin Kotschenreuther
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany
| | - Shuya Deng
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - David M Kofler
- Laboratory of Molecular Immunology, Division of Rheumatology and Clinical Immunology, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpenerstr. 62, 50937, Cologne, Germany.
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf, Cologne, Germany.
| |
Collapse
|
7
|
Orozco G, Gupta M, Gedaly R, Marti F. Untangling the Knots of Regulatory T Cell Therapy in Solid Organ Transplantation. Front Immunol 2022; 13:883855. [PMID: 35720387 PMCID: PMC9198594 DOI: 10.3389/fimmu.2022.883855] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022] Open
Abstract
Numerous preclinical studies have provided solid evidence supporting adoptive transfer of regulatory T cells (Tregs) to induce organ tolerance. As a result, there are 7 currently active Treg cell-based clinical trials in solid organ transplantation worldwide, all of which are early phase I or phase I/II trials. Although the results of these trials are optimistic and support both safety and feasibility, many experimental and clinical unanswered questions are slowing the progression of this new therapeutic alternative. In this review, we bring to the forefront the major challenges that Treg cell transplant investigators are currently facing, including the phenotypic and functional diversity of Treg cells, lineage stability, non-standardized ex vivo Treg cell manufacturing process, adequacy of administration route, inability of monitoring and tracking infused cells, and lack of biomarkers or validated surrogate endpoints of efficacy in clinical trials. With this plethora of interrogation marks, we are at a challenging and exciting crossroad where properly addressing these questions will determine the successful implementation of Treg cell-based immunotherapy in clinical transplantation.
Collapse
Affiliation(s)
- Gabriel Orozco
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Meera Gupta
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, United States.,Alliance Research Initiative [Treg cells to Induce Liver Tolerance (TILT) Alliance], University of Kentucky College of Medicine, Lexington, KY, United States
| | - Roberto Gedaly
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, United States.,Alliance Research Initiative [Treg cells to Induce Liver Tolerance (TILT) Alliance], University of Kentucky College of Medicine, Lexington, KY, United States.,Lucille Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, KY, United States
| | - Francesc Marti
- Department of Surgery - Transplant Division, College of Medicine, University of Kentucky, Lexington, KY, United States.,Alliance Research Initiative [Treg cells to Induce Liver Tolerance (TILT) Alliance], University of Kentucky College of Medicine, Lexington, KY, United States.,Lucille Parker Markey Cancer Center, University of Kentucky, College of Medicine, Lexington, KY, United States
| |
Collapse
|
8
|
Rezaei Kahmini F, Shahgaldi S, Azimi M, Mansourabadi AH. Emerging therapeutic potential of regulatory T (Treg) cells for rheumatoid arthritis: New insights and challenges. Int Immunopharmacol 2022; 108:108858. [PMID: 35597122 DOI: 10.1016/j.intimp.2022.108858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/27/2022] [Accepted: 05/10/2022] [Indexed: 11/05/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune-related disorder characterized by chronic inflammation. Although the etiopathogenesis of RA still remains to be clarified, it is supposed that the breakdown of immune self-tolerance may contribute to the development of RA. Thus, restoring of immune tolerance at the site of inflammation is the ultimate goal of RA treatment. Regulatory T cells (Treg cells) are the main suppressive cells that maintain tolerance and inhibit immunity against auto-antigen. Of note, recent studies demonstrated the efficacy of adoptive transfer of Treg cells in the modulation of the unwanted immune response, which makes them an ideal candidate to maintain immune homeostasis and restore antigen-specific tolerance in the case of RA and other autoimmune diseases. This review intends to submit recent finding of Treg cells-based therapies in RA with a focus on strategies applied to improve the therapeutic value of Treg cells to restore immune tolerance.
Collapse
Affiliation(s)
- Fatemeh Rezaei Kahmini
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Shahab Shahgaldi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Azimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Mansourabadi
- Department of Immunology, School of medicine, Tehran University of Medical Sciences, Tehran, Iran; Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
9
|
Harnessing the inherent power of chimeric antigen receptor (CAR)-expressing regulatory T cells (CAR-Tregs) to treat autoimmune-related disorders. Mol Biol Rep 2022; 49:4069-4078. [PMID: 35534581 DOI: 10.1007/s11033-022-07511-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
Over the past years, adoptive cell therapy with regulatory T lymphocytes (Tregs) has captured the attention of many scientists and clinicians as a novel promising approach for treating a wide range of immune-mediated disorders. In particular, the robust immunosuppressive properties of these cells have been demonstrated to make them uniquely valuable for the treatment of autoimmune diseases. More recently, it has been brought to light that adoptive transfer of chimeric antigen receptor (CAR) Tregs (CAR-Tregs) can also serve a protective role against autoimmune-related disorders. Interestingly, a growing body of evidence indicates that the beneficial and therapeutic effects of antigen-specific CAR-Tregs surpass those of polyclonal Tregs in treating autoimmune conditions. Therefore, harnessing and adapting CAR technology to generate more specific and effective CAR-Tregs, both in terms of tissue localization and antigen recognition, may lay the foundations for the development of far more potent immunotherapeutic strategies for autoimmune-related disorders. Herein, we first highlight the major immunosuppressive abilities of CAR-Tregs and further summarize the current findings on their potential applications in treating autoimmune-related disorders. Then, we will attempt to address the practical challenges in the clinical use of CAR-Treg therapies.
Collapse
|
10
|
Boardman DA, Levings MK. Emerging strategies for treating autoimmune disorders with genetically modified Treg cells. J Allergy Clin Immunol 2022; 149:1-11. [PMID: 34998473 DOI: 10.1016/j.jaci.2021.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022]
Abstract
Gene editing of living cells is a cornerstone of present-day medical research that has enabled scientists to address fundamental biologic questions and identify novel strategies to treat diseases. The ability to manipulate adoptive cell therapy products has revolutionized cancer immunotherapy and promises similar results for the treatment of autoimmune diseases, inflammatory disorders, and transplant rejection. Clinical trials have recently deemed polyclonal regulatory T (Treg) cell therapy to be a safe therapeutic option, but questions remain regarding the efficacy of this approach. In this review, we discuss how gene editing technologies are being applied to transform the future of Treg cell therapy, focusing on the preclinical strategies that are currently being investigated to enhance the efficacy, function, and survival of human Treg cells. We explore approaches that may be used to generate immunoregulatory cells ex vivo, detail emerging strategies that are being used to modify these cells (such as using chimeric antigen receptors to confer antigen specificity), and outline concepts that have been explored to repurpose conventional T cells to target and destroy autoreactive and alloreactive lymphocytes. We also describe the key hurdles that currently hinder the clinical adoption of Treg cell therapy and propose potential future avenues of research for this field.
Collapse
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
|
11
|
Park Y, Kwok SK. Recent Advances in Cell Therapeutics for Systemic Autoimmune Diseases. Immune Netw 2022; 22:e10. [PMID: 35291648 PMCID: PMC8901702 DOI: 10.4110/in.2022.22.e10] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/03/2022] Open
Abstract
Systemic autoimmune diseases arise from loss of self-tolerance and immune homeostasis between effector and regulator functions. There are many therapeutic modalities for autoimmune diseases ranging from conventional disease-modifying anti-rheumatic drugs and immunosuppressants exerting nonspecific immune suppression to targeted agents including biologic agents and small molecule inhibitors aiming at specific cytokines and intracellular signal pathways. However, such current therapeutic strategies can rarely induce recovery of immune tolerance in autoimmune disease patients. To overcome limitations of conventional treatment modalities, novel approaches using specific cell populations with immune-regulatory properties have been attempted to attenuate autoimmunity. Recently progressed biotechnologies enable sufficient in vitro expansion and proper manipulation of such ‘tolerogenic’ cell populations to be considered for clinical application. We introduce 3 representative cell types with immunosuppressive features, including mesenchymal stromal cells, Tregs, and myeloid-derived suppressor cells. Their cellular definitions, characteristics, mechanisms of immune regulation, and recent data about preclinical and clinical studies in systemic autoimmune diseases are reviewed here. Challenges and limitations of each cell therapy are also addressed.
Collapse
Affiliation(s)
- Youngjae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
12
|
CAR Treg: A new approach in the treatment of autoimmune diseases. Int Immunopharmacol 2021; 102:108409. [PMID: 34863655 DOI: 10.1016/j.intimp.2021.108409] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 12/22/2022]
Abstract
Regulatory T cells (Tregs) have the role of regulating self-tolerance, and suppressing immune responses. Defects in Treg function and number can lead to in loss of tolerance or autoimmune disease. To treat or control autoimmune diseases, one of the options is to develop immune tolerance for Tregs cell therapy, which includes promotion and activation. Recently, cell-based treatment as a promising approach to increase cells function and number has been developed. Cell therapy by chimeric T antigen receptor (CAR-T) cells has shown significant efficacy in the treatment of leukemia, which has led researchers to use CAR-T cells in other diseases like autoimmune diseases. Here, we describe the existing treatments for autoimmune diseases and the available treatments based on Treg, their benefits and restrictions for implementation in clinical trials. We also discussed potential solutions to overcome these limitations. It seems novel designs of CARs to be new hope for autoimmune diseases and expected to be a potential cure option in a wide array of disease in the future. Therefore, it is very important to address this issue and increase information about it.
Collapse
|
13
|
Selck C, Dominguez-Villar M. Antigen-Specific Regulatory T Cell Therapy in Autoimmune Diseases and Transplantation. Front Immunol 2021; 12:661875. [PMID: 34054826 PMCID: PMC8160309 DOI: 10.3389/fimmu.2021.661875] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/27/2021] [Indexed: 12/30/2022] Open
Abstract
Regulatory T (Treg) cells are a heterogenous population of immunosuppressive T cells whose therapeutic potential for the treatment of autoimmune diseases and graft rejection is currently being explored. While clinical trial results thus far support the safety and efficacy of adoptive therapies using polyclonal Treg cells, some studies suggest that antigen-specific Treg cells are more potent in regulating and improving immune tolerance in a disease-specific manner. Hence, several approaches to generate and/or expand antigen-specific Treg cells in vitro or in vivo are currently under investigation. However, antigen-specific Treg cell therapies face additional challenges that require further consideration, including the identification of disease-relevant antigens as well as the in vivo stability and migratory behavior of Treg cells following transfer. In this review, we discuss these approaches and the potential limitations and describe prospective strategies to enhance the efficacy of antigen-specific Treg cell treatments in autoimmunity and transplantation.
Collapse
Affiliation(s)
- Claudia Selck
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | | |
Collapse
|
14
|
Amini L, Greig J, Schmueck-Henneresse M, Volk HD, Bézie S, Reinke P, Guillonneau C, Wagner DL, Anegon I. Super-Treg: Toward a New Era of Adoptive Treg Therapy Enabled by Genetic Modifications. Front Immunol 2021; 11:611638. [PMID: 33717052 PMCID: PMC7945682 DOI: 10.3389/fimmu.2020.611638] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/24/2020] [Indexed: 12/27/2022] Open
Abstract
Regulatory Tcells (Treg) are essential components of peripheral immune homeostasis. Adoptive Treg cell therapy has shown efficacy in a variety of immune-mediated diseases in preclinical studies and is now moving from phase I/IIa to larger phase II studies aiming to demonstrate efficacy. However, hurdles such as in vivo stability and efficacy remain to be addressed. Nevertheless, preclinical models have shown that Treg function and specificity can be increased by pharmacological substances or gene modifications, and even that conventional T cells can be converted to Treg potentially providing new sources of Treg and facilitating Treg cell therapy. The exponential growth in genetic engineering techniques and their application to T cells coupled to a large body of knowledge on Treg open numerous opportunities to generate Treg with "superpowers". This review summarizes the genetic engineering techniques available and their applications for the next-generation of Super-Treg with increased function, stability, redirected specificity and survival.
Collapse
Affiliation(s)
- Leila Amini
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Jenny Greig
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Michael Schmueck-Henneresse
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Séverine Bézie
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Petra Reinke
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Carole Guillonneau
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Dimitrios L. Wagner
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ignacio Anegon
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
15
|
Regulatory T Cells for the Induction of Transplantation Tolerance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33523454 DOI: 10.1007/978-981-15-6407-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Organ transplantation is the optimal treatment for terminal and irreversible organ failure. Achieving transplantation tolerance has long been the ultimate goal in the field of transplantation. Regulatory T cell (Treg)-based therapy is a promising novel approach for inducing donor organ-specific tolerance. Tregs play critical roles in the maintenance of immune homeostasis and self-tolerance, by promoting transplantation tolerance through a variety of mechanisms on different target cells, including anti-inflammatory cytokine production, induction of apoptosis, disruption of metabolic pathways, and mutual interaction with dendritic cells. The continued success of Treg-based therapy in the clinical setting is critically dependent on preclinical studies that support its translational potential. However, although some initial clinical trials of adoptive Treg therapy have successively demonstrated safety and efficacy for immunosuppressant minimization and transplantation tolerance induction, most Treg-based hematopoietic stem cell and solid organ clinical trials are still in their infancy. These clinical trials have not only focused on safety and efficacy but also included optimization and standardization protocols of good manufacturing practice regarding cell isolation, expansion, dosing, timing, specificity, quality control, concomitant immunosuppressants, and post-administration monitoring. We herein report a brief introduction of Tregs, including their phenotypic and functional characterization, and focus on the clinical translation of Treg-based therapeutic applications in the setting of transplantation.
Collapse
|
16
|
Raffin C, Vo LT, Bluestone JA. T reg cell-based therapies: challenges and perspectives. Nat Rev Immunol 2020; 20:158-172. [PMID: 31811270 PMCID: PMC7814338 DOI: 10.1038/s41577-019-0232-6] [Citation(s) in RCA: 408] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2019] [Indexed: 12/25/2022]
Abstract
Cellular therapies using regulatory T (Treg) cells are currently undergoing clinical trials for the treatment of autoimmune diseases, transplant rejection and graft-versus-host disease. In this Review, we discuss the biology of Treg cells and describe new efforts in Treg cell engineering to enhance specificity, stability, functional activity and delivery. Finally, we envision that the success of Treg cell therapy in autoimmunity and transplantation will encourage the clinical use of adoptive Treg cell therapy for non-immune diseases, such as neurological disorders and tissue repair.
Collapse
Affiliation(s)
- Caroline Raffin
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Linda T Vo
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Eisenberg V, Hoogi S, Shamul A, Barliya T, Cohen CJ. T-cells "à la CAR-T(e)" - Genetically engineering T-cell response against cancer. Adv Drug Deliv Rev 2019; 141:23-40. [PMID: 30653988 DOI: 10.1016/j.addr.2019.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/01/2019] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
The last decade will be remembered as the dawn of the immunotherapy era during which we have witnessed the approval by regulatory agencies of genetically engineered CAR T-cells and of checkpoint inhibitors for cancer treatment. Understandably, T-lymphocytes represent the essential player in these approaches. These cells can mediate impressive tumor regression in terminally-ill cancer patients. Moreover, they are amenable to genetic engineering to improve their function and specificity. In the present review, we will give an overview of the most recent developments in the field of T-cell genetic engineering including TCR-gene transfer and CAR T-cells strategies. We will also elaborate on the development of other types of genetic modifications to enhance their anti-tumor immune response such as the use of co-stimulatory chimeric receptors (CCRs) and unconventional CARs built on non-antibody molecules. Finally, we will discuss recent advances in genome editing and synthetic biology applied to T-cell engineering and comment on the next challenges ahead.
Collapse
|
18
|
Optimizing regulatory T cells for therapeutic application in human organ transplantation. Curr Opin Organ Transplant 2018; 23:516-523. [DOI: 10.1097/mot.0000000000000561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
19
|
Okamura T, Yamamoto K, Fujio K. Early Growth Response Gene 2-Expressing CD4 +LAG3 + Regulatory T Cells: The Therapeutic Potential for Treating Autoimmune Diseases. Front Immunol 2018. [PMID: 29535721 PMCID: PMC5834469 DOI: 10.3389/fimmu.2018.00340] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Regulatory T cells (Tregs) are necessary for the maintenance of immune tolerance. Tregs are divided into two major populations: one is thymus derived and the other develops in the periphery. Among these Tregs, CD4+CD25+ Tregs, which mainly originate in the thymus, have been extensively studied. Transcription factor Foxp3 is well known as a master regulatory gene for the development and function of CD4+CD25+ Tregs. On the other hand, peripheral Tregs consist of distinct cell subsets including Foxp3-dependent extrathymically developed Tregs and interleukin (IL)-10-producing type I regulatory T (Tr1) cells. Lymphocyte activation gene 3 (LAG3) and CD49b are reliable cell surface markers for Tr1 cells. CD4+CD25−LAG3+ Tregs (LAG3+ Tregs) develop in the periphery and produce a large amount of IL-10. LAG3+ Tregs characteristically express the early growth response gene 2 (Egr2), a zinc-finger transcription factor, and exhibit its suppressive activity in a Foxp3-independent manner. Although Egr2 was known to be essential for hindbrain development and myelination of the peripheral nervous system, recent studies revealed that Egr2 plays vital roles in the induction of T cell anergy and also the suppressive activities of LAG3+ Tregs. Intriguingly, forced expression of Egr2 converts naive CD4+ T cells into IL-10-producing Tregs that highly express LAG3. Among the four Egr gene family members, Egr3 is thought to compensate for the function of Egr2. Recently, we reported that LAG3+ Tregs suppress humoral immune responses via transforming growth factor β3 production in an Egr2- and Egr3-dependent manner. In this review, we focus on the role of Egr2 in Tregs and also discuss its therapeutic potential for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan.,Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
Nowak A, Lock D, Bacher P, Hohnstein T, Vogt K, Gottfreund J, Giehr P, Polansky JK, Sawitzki B, Kaiser A, Walter J, Scheffold A. CD137+CD154- Expression As a Regulatory T Cell (Treg)-Specific Activation Signature for Identification and Sorting of Stable Human Tregs from In Vitro Expansion Cultures. Front Immunol 2018; 9:199. [PMID: 29467769 PMCID: PMC5808295 DOI: 10.3389/fimmu.2018.00199] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/23/2018] [Indexed: 01/30/2023] Open
Abstract
Regulatory T cells (Tregs) are an attractive therapeutic tool for several different immune pathologies. Therapeutic Treg application often requires prolonged in vitro culture to generate sufficient Treg numbers or to optimize their functionality, e.g., via genetic engineering of their antigen receptors. However, purity of clinical Treg expansion cultures is highly variable, and currently, it is impossible to identify and separate stable Tregs from contaminating effector T cells, either ex vivo or after prior expansion. This represents a major obstacle for quality assurance of expanded Tregs and raises significant safety concerns. Here, we describe a Treg activation signature that allows identification and sorting of epigenetically imprinted Tregs even after prolonged in vitro culture. We show that short-term reactivation resulted in expression of CD137 but not CD154 on stable FoxP3+ Tregs that displayed a demethylated Treg-specific demethylated region, high suppressive potential, and lack of inflammatory cytokine expression. We also applied this Treg activation signature for rapid testing of chimeric antigen receptor functionality in human Tregs and identified major differences in the signaling requirements regarding CD137 versus CD28 costimulation. Taken together, CD137+CD154- expression emerges as a universal Treg activation signature ex vivo and upon in vitro expansion allowing the identification and isolation of epigenetically stable antigen-activated Tregs and providing a means for their rapid functional testing in vitro.
Collapse
Affiliation(s)
- Anna Nowak
- German Rheumatism Research Centre (DRFZ) Berlin, Leibniz Association, Berlin, Germany
| | - Dominik Lock
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Petra Bacher
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University Medicine, Berlin, Germany
| | - Thordis Hohnstein
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University Medicine, Berlin, Germany
| | - Katrin Vogt
- Institute for Medical Immunology, Charité - University Medicine, Berlin, Germany
| | - Judith Gottfreund
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Pascal Giehr
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Julia K Polansky
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine, Berlin, Germany
| | - Birgit Sawitzki
- Institute for Medical Immunology, Charité - University Medicine, Berlin, Germany
| | | | - Jörn Walter
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Alexander Scheffold
- German Rheumatism Research Centre (DRFZ) Berlin, Leibniz Association, Berlin, Germany.,Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - University Medicine, Berlin, Germany
| |
Collapse
|
21
|
Vaikunthanathan T, Safinia N, Boardman D, Lechler RI, Lombardi G. Regulatory T cells: tolerance induction in solid organ transplantation. Clin Exp Immunol 2017; 189:197-210. [PMID: 28422316 DOI: 10.1111/cei.12978] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
The concept of regulatory T cell (Treg ) therapy in transplantation is now a reality. Significant advances in science and technology have enabled us to isolate human Tregs , expand them to clinically relevant numbers and infuse them into human transplant recipients. With several Phase I/II trials under way investigating Treg safety and efficacy it is now more crucial than ever to understand their complex biology. However, our journey is by no means complete; results from these trials will undoubtedly provoke both further knowledge and enquiry which, alongside evolving science, will continue to drive the optimization of Treg therapy in the pursuit of transplantation tolerance. In this review we will summarize current knowledge of Treg biology, explore novel technologies in the setting of Treg immunotherapy and address key prerequisites surrounding the clinical application of Tregs in transplantation.
Collapse
Affiliation(s)
- T Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - N Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - D Boardman
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - R I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - G Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| |
Collapse
|
22
|
Antigen-specificity using chimeric antigen receptors: the future of regulatory T-cell therapy? Biochem Soc Trans 2016; 44:342-8. [DOI: 10.1042/bst20150247] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 12/24/2022]
Abstract
Adoptive regulatory T-cell (Treg) therapy using autologous Tregs expanded ex vivo is a promising therapeutic approach which is currently being investigated clinically as a means of treating various autoimmune diseases and transplant rejection. Despite this, early results have highlighted the need for potent Tregs to yield a substantial clinical advantage. One way to achieve this is to create antigen-specific Tregs which have been shown in pre-clinical animal models to have an increased potency at suppressing undesired immune responses, compared to polyclonal Tregs. This mini review outlines where Treg therapy currently stands and discusses the approaches which may be taken to generate antigen-specific Tregs, including the potential use of chimeric antigen receptors (CARs), for future clinical trials.
Collapse
|
23
|
Ishigaki K, Shoda H, Kochi Y, Yasui T, Kadono Y, Tanaka S, Fujio K, Yamamoto K. Quantitative and qualitative characterization of expanded CD4+ T cell clones in rheumatoid arthritis patients. Sci Rep 2015; 5:12937. [PMID: 26245356 PMCID: PMC4542667 DOI: 10.1038/srep12937] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/06/2015] [Indexed: 12/04/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune destructive arthritis associated with CD4+ T cell-mediated immunity. Although expanded CD4+ T cell clones (ECs) has already been confirmed, the detailed characteristics of ECs have not been elucidated in RA. Using combination of a single-cell analysis and next-generation sequencing (NGS) in TCR repertoire analysis, we here revealed the detailed nature of ECs by examining peripheral blood (PB) from 5 RA patients and synovium from 1 RA patient. When we intensively investigated the single-cell transcriptome of the most expanded clones in memory CD4+ T cells (memory-mECs) in RA-PB, senescence-related transcripts were up-regulated, indicating circulating ECs were constantly stimulated. Tracking of the transcriptome shift within the same memory-mECs between PB and the synovium revealed the augmentations in senescence-related gene expression and the up-regulation of synovium-homing chemokine receptors in the synovium. Our in-depth characterization of ECs in RA successfully demonstrated the presence of the specific immunological selection pressure, which determines the phenotype of ECs. Moreover, transcriptome tracking added novel aspects to the underlying sequential immune processes. Our approach may provide new insights into the pathophysiology of RA.
Collapse
Affiliation(s)
- Kazuyoshi Ishigaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Tetsuro Yasui
- Department of Orthopaedic Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Yuho Kadono
- Department of Orthopaedic Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
24
|
Fujio K, Okamura T, Okamoto A, Yamamoto K. T-cell receptor- and anti-inflammatory gene-modulated T cells as therapy for autoimmune disease. Expert Rev Clin Immunol 2014; 3:883-90. [DOI: 10.1586/1744666x.3.6.883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Wright GP, Ehrenstein MR, Stauss HJ. Regulatory T-cell adoptive immunotherapy: potential for treatment of autoimmunity. Expert Rev Clin Immunol 2014; 7:213-25. [DOI: 10.1586/eci.10.96] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Jethwa H, Adami AA, Maher J. Use of gene-modified regulatory T-cells to control autoimmune and alloimmune pathology: is now the right time? Clin Immunol 2013; 150:51-63. [PMID: 24333533 DOI: 10.1016/j.clim.2013.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/21/2013] [Accepted: 11/06/2013] [Indexed: 12/22/2022]
Abstract
Adoptive immunotherapy using genetically targeted T-cells has recently begun to achieve impressive clinical impact in selected tumor types. Furthermore, long-term follow-up studies indicate thus far that integrating viral vectors do not elicit clinically evident genotoxicity in T-cells, unlike hematopoietic stem cells. The optimism engendered by this clinical experience provides a platform for consideration of the extended use of this technology in other disease types. One area of particular interest entails the harnessing of regulatory T-cells (Tregs) in order to down-regulate unwanted immune responses. Increasing evidence supports the efficacy of this approach in pre-clinical models of autoimmune disease and allograft rejection. Nonetheless, questions remain about optimal host cell, transgene cargo, phenotypic stability of engineered cells in vivo and potential for toxicity. Here, we review the evidence that genetically engineered Tregs can effectively dampen pathogenic immune responses and critically evaluate the prospects for clinical development of this approach.
Collapse
Affiliation(s)
- Hannah Jethwa
- Department of Medicine, Barnet and Chase Farm NHS Trust, Barnet, Hertfordshire EN5 3DJ, UK
| | - Antonella A Adami
- King's College London, King's Health Partners Integrated Cancer Centre, Department of Research Oncology, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK
| | - John Maher
- King's College London, King's Health Partners Integrated Cancer Centre, Department of Research Oncology, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK; Department of Immunology, Barnet and Chase Farm NHS Trust, Barnet, Hertfordshire EN5 3DJ, UK; Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK.
| |
Collapse
|
27
|
Sumitomo S, Fujio K, Okamura T, Morita K, Ishigaki K, Suzukawa K, Kanaya K, Kondo K, Yamasoba T, Furukawa A, Kitahara N, Shoda H, Shibuya M, Okamoto A, Yamamoto K. Transcription factor early growth response 3 is associated with the TGF-β1 expression and the regulatory activity of CD4-positive T cells in vivo. THE JOURNAL OF IMMUNOLOGY 2013; 191:2351-9. [PMID: 23904169 DOI: 10.4049/jimmunol.1202106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TGF-β1 is an important anti-inflammatory cytokine, and several regulatory T cell (Treg) subsets including CD4(+)CD25(+)Foxp3(+) Tregs and Th3 cells have been reported to exert regulatory activity via the production of TGF-β1. However, it has not yet been elucidated which transcription factor is involved in TGF-β1 transcription. Early growth response 3 (Egr-3) is a zinc-finger transcription factor that creates and maintains T cell anergy. In this study, we found that Egr-3 induces the expression of TGF-β1 in both murine and human CD4(+) T cells. Egr-3 overexpression in murine CD4(+) T cells induced the production of TGF-β1 and enhanced the phosphorylation of STAT3, which is associated with TGF-β1 transcription. Moreover, Egr-3 conferred Ag-specific regulatory activity on murine CD4(+) T cells. In collagen-induced arthritis and delayed-type hypersensitivity model mice, Egr-3-transduced CD4(+) T cells exhibited significant regulatory activity in vivo. In particular, the suppression of delayed-type hypersensitivity depended on TGF-β1. In human tonsils, we found that CD4(+)CD25(-)CD45RO(-)lymphocyte activation gene 3 (LAG3)(-) T cells express membrane-bound TGF-β1 in an EGR3-dependent manner. Gene-expression analysis revealed that CD4(+)CD25(-)CD45RO(-)LAG3(-) T cells are quite different from conventional CD4(+)CD25(+)Foxp3(+) Tregs. Intriguingly, the CD4(+)CD25(-)CD45RO(-)LAG3(-) T cells suppressed graft-versus-host disease in immunodeficient mice transplanted with human PBMCs. Our results suggest that Egr-3 is a transcription factor associated with TGF-β1 expression and in vivo regulatory activity in both mice and humans.
Collapse
Affiliation(s)
- Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Daniel-Meshulam I, Ya'akobi S, Ankri C, Cohen CJ. How (specific) would like your T-cells today? Generating T-cell therapeutic function through TCR-gene transfer. Front Immunol 2012; 3:186. [PMID: 22783259 PMCID: PMC3390604 DOI: 10.3389/fimmu.2012.00186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/15/2012] [Indexed: 01/02/2023] Open
Abstract
T-cells are central players in the immune response against both pathogens and cancer. Their specificity is solely dictated by the T-cell receptor (TCR) they clonally express. As such, the genetic modification of T lymphocytes using pathogen- or cancer-specific TCRs represents an appealing strategy to generate a desired immune response from peripheral blood lymphocytes. Moreover, notable objective clinical responses were observed in terminally ill cancer patients treated with TCR-gene modified cells in several clinical trials conducted recently. Nevertheless, several key aspects of this approach are the object of intensive research aimed at improving the reliability and efficacy of this strategy. Herein, we will survey recent studies in the field of TCR-gene transfer dealing with the improvement of this approach and its application for the treatment of malignant, autoimmune, and infectious diseases.
Collapse
Affiliation(s)
- Inbal Daniel-Meshulam
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences , Bar-Ilan University, Ramat Gan, Israel
| | | | | | | |
Collapse
|
30
|
Evaluation of Epstein-Barr virus latent membrane protein 2 specific T-cell receptors driven by T-cell specific promoters using lentiviral vector. Clin Dev Immunol 2011; 2011:716926. [PMID: 21969838 PMCID: PMC3182378 DOI: 10.1155/2011/716926] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 07/26/2011] [Accepted: 07/26/2011] [Indexed: 02/05/2023]
Abstract
Transduction of latent membrane protein 2 (LMP2)-specific T-cell receptors into activated T lymphocytes may provide a universal, MHC-restricted mean to treat EBV-associated tumors in adoptive immunotherapy. We compared TCR-specific promoters of distinct origin in lentiviral vectors, that is, Vβ6.7, delta, luria, and Vβ5.1 to evaluate TCR gene expression in human primary peripheral blood monocytes and T cell line HSB2. Vectors containing Vβ 6.7 promoter were found to be optimal for expression in PBMCs, and they maintained expression of the transduced TCRs for up to 7 weeks. These cells had the potential to recognize subdominant EBV latency antigens as measured by cytotoxicity and IFN-γ secretion. The nude mice also exhibited significant resistance to the HLA-A2 and LMP2-positive CNE tumor cell challenge after being infused with lentiviral transduced CTLs. In conclusion, LMP2-specific CTLs by lentiviral transduction have the potential use for treatment of EBV-related tumors.
Collapse
|
31
|
Resistance to regulatory T cell-mediated suppression in rheumatoid arthritis can be bypassed by ectopic foxp3 expression in pathogenic synovial T cells. Proc Natl Acad Sci U S A 2011; 108:16717-22. [PMID: 21926327 DOI: 10.1073/pnas.1112722108] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Increasing evidence suggests that regulatory T cell (Treg) function is impaired in chronic inflammatory diseases such as rheumatoid arthritis (RA). Here we demonstrate that Tregs are unable to modulate the spontaneous production of TNF-α from RA synovial cells cultured from the diseased synovium site. Cytokine (IL-2, IL-6, TNF-α) activated T cells (Tck), cells we previously demonstrated to mimic the effector function of pathogenic RA synovial T cells, contained Tregs that survived and divided in this cytokine environment; however, the up-regulation of key molecules associated with Treg function (CTLA-4 and LFA-1) was impaired. Furthermore, Tregs were unable to suppress the function of Tcks, including contact-dependent induction of TNF-α from macrophages, supporting the concept that impaired Treg function/responsiveness contributes to chronicity of RA. However, ectopic foxp3 expression in both Tcks and pathogenic RA synovial T cells attenuated their cytokine production and function, including contact-dependent activation of macrophages. This diminished response to cytokine activation after ectopic foxp3 expression involved inhibited NF-κB activity and differed mechanistically from that displayed endogenously in conventional Tregs. These results suggest that diseases such as RA may perpetuate owing to the inability of Tregs to control cytokine-activated T-cell function. Understanding the mechanism whereby foxp3 attenuates the pathogenic function of synovial T cells may provide insight into the mechanisms of chronicity in inflammatory disease and potentially reveal new therapeutic candidates.
Collapse
|
32
|
Okamoto A, Fujio K, Yamamoto K. The future of lupus therapy modulating autoantigen recognition. Lupus 2011; 19:1474-81. [PMID: 20947560 DOI: 10.1177/0961203310374306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The mainstay of the current treatment for systemic lupus erythematosus consists of steroids and immunosuppressants. However, these non-specific immunosuppressive therapies can cause infection and other serious adverse events. The regulation of the autoantigen-specific immune response is a promising therapeutic approach with maximal efficacy and minimal adverse effects. T cells are essential components of antigen-specificity in the immune system. At present, we do not have a sufficient strategy for manipulating the responses of antigen-specific T cells. In this review, we describe the efficacy of two therapeutic approaches involving the modulation of autoantigen recognition by T cells in lupus model mice: (1) therapy involving engineered autoantigen-specific regulatory T cells generated by the gene transfer of autoantigen-specific TCR genes and appropriate regulatory genes into self lymphocytes; (2) therapy involving selective depletion of autoantigen presenting phagocytes. These selective immunosuppressive approaches could be useful strategies for the treatment of systemic lupus erythematosus.
Collapse
Affiliation(s)
- A Okamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
33
|
Haque R, Lei F, Xiong X, Wu Y, Song J. FoxP3 and Bcl-xL cooperatively promote regulatory T cell persistence and prevention of arthritis development. Arthritis Res Ther 2010; 12:R66. [PMID: 20384988 PMCID: PMC2888221 DOI: 10.1186/ar2983] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 02/16/2010] [Accepted: 04/12/2010] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Forkhead box p3 (FoxP3)-expressing regulatory T cells (Tregs) have been clearly implicated in the control of autoimmune disease in murine models. In addition, ectopic expression of FoxP3 conveys a Treg phenotype to CD4(+) T cells, lending itself to therapeutic use in the prevention of rheumatoid arthritis (RA). In this study, we generated therapeutically active Tregs with an increased life span and hence greater therapeutic potential. METHODS We used retrovirus-mediated transduction to introduce FoxP3 or FoxP3 with anti-apoptotic Bcl-2 family molecule Bcl-xL linked by a 2A picornavirus self-cleaving peptide into CD4(+) T cells to generate Tregs. In addition, by using in vitro functional analyses and adoptive immunotherapy in a murine model of RA, we demonstrated that these Tregs were highly reactive. RESULTS We found that CD4(+) T cells expressing both FoxP3 and Bcl-xL were able to differentiate into functional Tregs, which have a long-term survival advantage over cells transduced with FoxP3 alone. In an in vivo murine model, adoptive transfer of Tregs expressing both FoxP3 and Bcl-xL demonstrated more effective suppression of RA than CD4(+) T cells expressing FoxP3 alone. CONCLUSIONS FoxP3 and Bcl-xL can cooperatively promote the differentiation and persistence of Tregs, with the capacity to prevent arthritis. Our results provide a novel approach for generating highly reactive Tregs for augmenting cellular immunotherapy for autoimmune disease.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/prevention & control
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/prevention & control
- Forkhead Transcription Factors/genetics
- Immunotherapy, Adoptive
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Transduction, Genetic
- bcl-X Protein/genetics
Collapse
Affiliation(s)
- Rizwanul Haque
- Department of Microbiology & Immunology and Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Fengyang Lei
- Department of Microbiology & Immunology and Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Xiaofang Xiong
- Department of Microbiology & Immunology and Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Yuzhang Wu
- Institute of Immunology, The Third Military Medical University, 30 Gaotanyan Street, Chongqing 400038, PR China
| | - Jianxun Song
- Department of Microbiology & Immunology and Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
- Institute of Immunology, The Third Military Medical University, 30 Gaotanyan Street, Chongqing 400038, PR China
| |
Collapse
|
34
|
Wright GP, Notley CA, Xue SA, Bendle GM, Holler A, Schumacher TN, Ehrenstein MR, Stauss HJ. Adoptive therapy with redirected primary regulatory T cells results in antigen-specific suppression of arthritis. Proc Natl Acad Sci U S A 2009; 106:19078-83. [PMID: 19884493 PMCID: PMC2776462 DOI: 10.1073/pnas.0907396106] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) can suppress a wide range of immune cells, making them an ideal candidate for the treatment of autoimmunity. The potential clinical translation of targeted therapy with antigen-specific Tregs is hampered by the difficulties of isolating rare specificities from the natural polyclonal T cell repertoire. Moreover, the initiating antigen is often unknown in autoimmune disease. Here we tested the ability of antigen-specific Tregs generated by retroviral gene transfer to ameliorate arthritis through linked suppression and therefore without cognate recognition of the disease-initiating antigen. We explored two distinct strategies: T cell receptor (TCR) gene transfer into purified CD4+CD25+ T cells was used to redirect the specificity of naturally occurring Tregs; and co-transfer of FoxP3 and TCR genes served to convert conventional CD4(+) T cells into antigen-specific regulators. Following adoptive transfer into recipient mice, the gene-modified T cells engrafted efficiently and retained TCR and FoxP3 expression. Using an established arthritis model, we demonstrate antigen-driven accumulation of the gene modified T cells at the site of joint inflammation, which resulted in a local reduction in the number of inflammatory Th17 cells and a significant decrease in arthritic bone destruction. Together, we describe a robust strategy to rapidly generate antigen-specific regulatory T cells capable of highly targeted inhibition of tissue damage in the absence of systemic immune suppression. This opens the possibility to target Tregs to tissue-specific antigens for the treatment of autoimmune tissue damage without the knowledge of the disease-causing autoantigens recognized by pathogenic T cells.
Collapse
Affiliation(s)
- Graham P. Wright
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| | - Clare A. Notley
- Department of Medicine, Centre for Rheumatology, University College London, London W1T 4JF, United Kingdom; and
| | - Shao-An Xue
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| | - Gavin M. Bendle
- Netherlands Cancer Institute, Department of Immunology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Angelika Holler
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| | - Ton N. Schumacher
- Netherlands Cancer Institute, Department of Immunology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Michael R. Ehrenstein
- Department of Medicine, Centre for Rheumatology, University College London, London W1T 4JF, United Kingdom; and
| | - Hans J. Stauss
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| |
Collapse
|
35
|
Okamura T, Fujio K, Shibuya M, Sumitomo S, Shoda H, Sakaguchi S, Yamamoto K. CD4+CD25-LAG3+ regulatory T cells controlled by the transcription factor Egr-2. Proc Natl Acad Sci U S A 2009; 106:13974-9. [PMID: 19666526 PMCID: PMC2729005 DOI: 10.1073/pnas.0906872106] [Citation(s) in RCA: 173] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Indexed: 12/15/2022] Open
Abstract
Regulatory T cells (Tregs) are engaged in the maintenance of immunological self-tolerance and immune homeostasis. IL-10 has an important role in maintaining the normal immune state. Here, we show that IL-10-secreting Tregs can be delineated in normal mice as CD4(+)CD25(-)Foxp3(-) T cells that express lymphocyte activation gene 3 (LAG-3), an MHC-class-II-binding CD4 homolog. Although approximately 2% of the CD4(+)CD25(-) T cell population consisted of CD4(+)CD25(-)LAG3(+) T cells in the spleen, CD4(+)CD25(-)LAG3(+) T cells are enriched to approximately 8% in the Peyer's patch. They are hypoproliferative upon in vitro antigenic stimulation and suppress in vivo development of colitis. Gene expression analysis reveals that CD4(+)CD25(-)LAG3(+) Tregs characteristically express early growth response gene 2 (Egr-2), a key molecule for anergy induction. Retroviral gene transfer of Egr-2 converts naïve CD4(+) T cells into the IL-10-secreting and LAG-3-expressing phenotype, and Egr-2-transduced CD4(+) T cells exhibit antigen-specific immunosuppressive capacity in vivo. Unlike Foxp3(+) natural Tregs, high-affinity interactions with selecting peptide/MHC ligands expressed in the thymus do not induce the development of CD4(+)CD25(-)LAG3(+) Tregs. In contrast, the number of CD4(+)CD25(-)LAG3(+) Tregs is influenced by the presence of environmental microbiota. Thus, IL-10-secreting Egr-2(+)LAG3(+)CD4(+) Tregs can be exploited for the control of peripheral immunity.
Collapse
Affiliation(s)
- Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and
| | - Mihoko Shibuya
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and
| | - Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and
| | - Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; and
| |
Collapse
|
36
|
Andersen KG, Butcher T, Betz AG. Specific immunosuppression with inducible Foxp3-transduced polyclonal T cells. PLoS Biol 2008; 6:e276. [PMID: 18998771 PMCID: PMC2581628 DOI: 10.1371/journal.pbio.0060276] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Accepted: 09/29/2008] [Indexed: 12/31/2022] Open
Abstract
Forkhead box p3 (Foxp3)-expressing regulatory T cells are key mediators of peripheral tolerance suppressing undesirable immune responses. Ectopic expression of Foxp3 confers regulatory T cell phenotype to conventional T cells, lending itself to therapeutic use in the prevention of autoimmunity and transplant rejection. Here, we show that adoptive transfer of polyclonal, wild-type T cells transduced with an inducible form of Foxp3 (iFoxp3) can be used to suppress immune responses on demand. In contrast to Foxp3-transduced cells, iFoxp3-transduced cells home "correctly" into secondary lymphoid organs, where they expand and participate in immune responses. Upon induction of iFoxp3, the cells assume regulatory T cell phenotype and start to suppress the response they initially partook in without causing systemic immunosuppression. We used this approach to suppress collagen-induced arthritis, in which conventional Foxp3-transduced cells failed to show any effect. This provides us with a generally applicable strategy to specifically halt immune responses on demand without prior knowledge of the antigens involved.
Collapse
Affiliation(s)
- Kristian G Andersen
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Tracey Butcher
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Alexander G Betz
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
37
|
Delgado M, Toscano MG, Benabdellah K, Cobo M, O'Valle F, Gonzalez-Rey E, Martín F. In vivo delivery of lentiviral vectors expressing vasoactive intestinal peptide complementary DNA as gene therapy for collagen-induced arthritis. ACTA ACUST UNITED AC 2008; 58:1026-37. [PMID: 18383372 DOI: 10.1002/art.23283] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Vasoactive intestinal peptide (VIP) has been shown to exert potent immunomodulatory activity, and the use of lentiviral vectors has been found to be an effective means of gene delivery. The present study was therefore undertaken to investigate the feasibility and efficiency of gene therapy using lentiviral vectors expressing VIP (LentiVIP) for the treatment of rheumatoid arthritis (RA). METHODS We evaluated the therapeutic potential of the gene therapy strategy in the collagen-induced arthritis (CIA) mouse model, administering the vectors at different phases of the disease. The inflammatory response was determined by measuring the levels of various inflammatory cytokines and chemokines in the joints and serum. The Th1-mediated response was evaluated by determining the proliferative response and cytokine profile of T cells stimulated with autoantigen. RESULTS A single intraperitoneal injection of LentiVIP was highly effective in treating CIA. Mice with established, severe arthritis showed complete regression of the disease. The therapeutic effect of LentiVIP was associated with widespread biodistribution of the vector and increased VIP levels, especially in joints and lymphoid organs, and was mediated through a striking reduction of the 2 deleterious components of the disease, i.e., the autoimmune response (self-reactive Th1 cell activity and autoantibody production) and the inflammatory response. LentiVIP treatment also induced the generation and/or activation of CD4+,CD25+,FoxP3+ Treg cells in arthritic mice. CONCLUSION Our findings show that in vivo administration of lentiviral vector expressing VIP produces one of the most potent therapeutic effects described so far in any animal model of RA. We propose that VIP gene transfer should be further investigated as a potential novel, effective treatment of RA and other chronic autoimmune disorders.
Collapse
Affiliation(s)
- Mario Delgado
- Immunology and Cell Biology Department, Instituto de Parasitología y Biomedicina López Neyra, CSIC, Granada, Spain
| | | | | | | | | | | | | |
Collapse
|
38
|
Fujio K, Okamura T, Okamoto A, Yamamoto K. T Cell Receptor Gene Therapy for Autoimmune Diseases. Ann N Y Acad Sci 2007; 1110:222-32. [PMID: 17911437 DOI: 10.1196/annals.1423.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The current quality of autoimmune disease treatments is not satisfactory in regard to efficacy and safety. Antigen-specific immunotherapy is a future therapy that could achieve maximal efficacy with minimal adverse effects. T cells are essential components in antigen-specific immunity. However, we do not have a sufficient strategy for manipulating antigen-specific T cells. We propose that T cell receptor (TCR) gene transfer is a hopeful approach for antigen-specific immunotherapy. We confirmed the efficacy of TCR gene therapy in animal models of systemic autoimmune disease and arthritis. In lupus-prone NZB/W F1 mice, nucleosome-specific TCR and CTLA4Ig transduced cells suppressed autoantibody production and nephritis development. In the therapeutic experiment of collagen-induced arthritis (CIA), arthritis-related TCRs were isolated from single T cells accumulating in the arthritis site. Arthritis-related TCR and TNFRIg transduced cells or TCR and Foxp3 transduced cells suppressed arthritis progression and bone destruction. Therefore, engineered antigen-specific cells manipulated to express appropriate functional genes could be applied to specific immunotherapy.
Collapse
Affiliation(s)
- Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113, Japan.
| | | | | | | |
Collapse
|
39
|
Abstract
The status of autoimmune disease therapies is not satisfactory. Antigen-specific immunotherapy has potential as a future therapy that could deliver maximal efficacy with minimal adverse effects. Several trials of antigen-specific immunotherapy have been performed, but so far no clear directions have been established. With regard to antigen-specificity in the immune system, T cells are essential components. However, at present, we do not have a sufficient range of strategies for manipulating antigen-specific T cells. In this review, the authors propose that T cell receptor gene transfer could be used for antigen-specific immunotherapy. In the proposed technique, important disease-related and, thus, antigen-specific T cells in patients would first be identified, and then a pair of cDNAs encoding alpha and beta T cell receptors would be isolated from these single T cells. These genes would then be transferred into self lymphocytes. These engineered antigen-specific cells can also be manipulated to express appropriate functional genes that could then be applied to specific immunotherapy.
Collapse
Affiliation(s)
- Kazuhiko Yamamoto
- University of Tokyo, Department of Allergy and Rheumatology, Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113, Japan.
| | | | | |
Collapse
|
40
|
Richman SA, Kranz DM. Display, engineering, and applications of antigen-specific T cell receptors. ACTA ACUST UNITED AC 2007; 24:361-73. [PMID: 17409021 DOI: 10.1016/j.bioeng.2007.02.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 02/23/2007] [Accepted: 02/26/2007] [Indexed: 10/23/2022]
Abstract
The use of T cell receptors (TCRs) as potential therapeutic agents provides an opportunity to target a greatly expanded array of antigens, compared to those now targeted with monoclonal antibodies. With the advent of new display technologies and TCR formats for in vitro engineering, it should be possible to generate high-affinity TCRs against virtually any peptide antigen that is shown to bind to a major histocompatibility complex (MHC) molecule (e.g. peptides derived from viral antigens or from self proteins that are associated with the transformed phenotype). What remains, however, are challenges associated with effective targeting of very low numbers of cell surface antigens (pepMHC), fewer than the case for conventional monoclonal antibody-based therapies. This hurdle might be overcome with the attachment of more effective payloads for soluble TCR approaches, or by using TCR gene transfer into T cells that can then be adoptively transferred into patients. There is considerable work to be done on the physiological aspects of either approach, including pharmacokinetic studies in the case of soluble TCRs, and T cell trafficking, persistence, and autoreactivity studies in the case of adoptively transferred T cells. As with the field of monoclonal antibodies, it will take time to explore these issues, but the potential benefits of TCR-based therapies make these challenges worth the effort.
Collapse
Affiliation(s)
- Sarah A Richman
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | | |
Collapse
|