101
|
Copsel S, Wolf D, Komanduri KV, Levy RB. The promise of CD4 +FoxP3 + regulatory T-cell manipulation in vivo: applications for allogeneic hematopoietic stem cell transplantation. Haematologica 2019; 104:1309-1321. [PMID: 31221786 PMCID: PMC6601084 DOI: 10.3324/haematol.2018.198838] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
CD4+FoxP3+ regulatory T cells (Tregs) are a non-redundant population critical for the maintenance of self-tolerance. Over the past decade, the use of these cells for therapeutic purposes in transplantation and autoimmune disease has emerged based on their capacity to inhibit immune activation. Basic science discoveries have led to identifying key receptors on Tregs that can regulate their proliferation and function. Notably, the understanding that IL-2 signaling is crucial for Treg homeostasis promoted the hypothesis that in vivo IL-2 treatment could provide a strategy to control the compartment. The use of low-dose IL-2 in vivo was shown to selectively expand Tregs versus other immune cells. Interestingly, a number of other Treg cell surface proteins, including CD28, CD45, IL-33R and TNFRSF members, have been identified which can also induce activation and proliferation of this population. Pre-clinical studies have exploited these observations to prevent and treat mice developing autoimmune diseases and graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation. These findings support the development of translational strategies to expand Tregs in patients. Excitingly, the use of low-dose IL-2 for patients suffering from graft-versus-host disease and autoimmune disease has demonstrated increased Treg levels together with beneficial outcomes. To date, promising pre-clinical and clinical studies have directly targeted Tregs and clearly established the ability to increase their levels and augment their function in vivo. Here we review the evolving field of in vivo Treg manipulation and its application to allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Krishna V Komanduri
- Department of Microbiology and Immunology.,Sylvester Comprehensive Cancer Center.,Division of Transplantation and Cellular Therapy, Department of Medicine
| | - Robert B Levy
- Department of Microbiology and Immunology .,Division of Transplantation and Cellular Therapy, Department of Medicine.,Department of Ophthalmology, Miller School of Medicine, University of Miami, FL, USA
| |
Collapse
|
102
|
Abstract
With the advent of the concept of dominant tolerance and the subsequent discovery of CD4+ regulatory T cells expressing the transcription factor FOXP3 (Tregs), almost all productive as well as nonproductive immune responses can be compartmentalized to a binary of immune effector T cells and immune regulatory Treg populations. A beneficial immune response warrants the timely regulation by Tregs, whereas a nonproductive immune response indicates insufficient effector functions or an outright failure of tolerance. There are ample reports supporting role of Tregs in suppressing spontaneous auto-immune diseases as well as promoting immune evasion by cancers. To top up their importance, several non-immune functions like tissue homeostasis and regeneration are also being attributed to Tregs. Hence, after being in the center stage of basic and translational immunological research, Tregs are making the next jump towards clinical studies. Therefore, newer small molecules, biologics as well as adoptive cell therapy (ACT) approaches are being tested to augment or undermine Treg responses in the context of autoimmunity and cancer. In this brief review, we present the strategies to modulate Tregs towards a favorable clinical outcome.
Collapse
Affiliation(s)
- Amit Sharma
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS) , Pohang , Republic of Korea.,Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology (POSTECH) , Pohang , Republic of Korea
| | - Dipayan Rudra
- Academy of Immunology and Microbiology, Institute for Basic Science (IBS) , Pohang , Republic of Korea.,Division of Integrative Biosciences & Biotechnology, Pohang University of Science and Technology (POSTECH) , Pohang , Republic of Korea
| |
Collapse
|
103
|
Yi L, Weifan Y, Huan Y. Chimeric antigen receptor-engineered regulatory T lymphocytes: promise for immunotherapy of autoimmune disease. Cytotherapy 2019; 21:925-934. [PMID: 31105041 DOI: 10.1016/j.jcyt.2019.04.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 02/01/2019] [Accepted: 04/28/2019] [Indexed: 02/06/2023]
Abstract
Regulatory T lymphocytes (Tregs) exist as natural ideal immunosuppressors in the immune system. Autologous or allogeneic Treg transfusion therapy has been carried out in animal models and humans as a new strategy for treating autoimmune disease. Recent studies have shown that Tregs can be engineered with chimeric antigen receptors to be antigen-specific, which are more effective than polyclonal Tregs with fewer target limitations and a lack of major histocompatibility complex restriction. This review describes the potential for applying chimeric antigen receptor-engineered regulatory T cells in autoimmune diseases.
Collapse
Affiliation(s)
- Li Yi
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yin Weifan
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yang Huan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
104
|
MacDonald KN, Piret JM, Levings MK. Methods to manufacture regulatory T cells for cell therapy. Clin Exp Immunol 2019; 197:52-63. [PMID: 30913302 DOI: 10.1111/cei.13297] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2019] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cell (Treg ) therapy has shown promise in early clinical trials for treating graft-versus-host disease, transplant rejection and autoimmune disorders. A challenge has been to isolate sufficiently pure Tregs and expand them to a clinical dose. However, there has been considerable progress in the development and optimization of these methods, resulting in a variety of manufacturing protocols being tested in clinical trials. In this review, we summarize methods that have been used to manufacture Tregs for clinical trials, including the choice of cell source and protocols for cell isolation and expansion. We also discuss alternative culture or genome editing methods for modulating Treg specificity, function or stability that could be applied to future clinical manufacturing protocols to increase the efficacy of Treg therapy.
Collapse
Affiliation(s)
- K N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - J M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
| | - M K Levings
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
105
|
Abstract
De novo donor-specific antibody (DSA) formation is a major problem in transplantation, and associated with long-term graft decline and loss as well as sensitization, limiting future transplant options. Forming high-affinity, long-lived antibody responses involves a process called the germinal center (GC) reaction, and requires interaction between several cell types, including GC B cells, T follicular helper (Tfh) and T follicular regulatory (Tfr) cells. T follicular regulatory cells are an essential component of the GC reaction, limiting its size and reducing nonspecific or self-reactive responses.An imbalance between helper function and regulatory function can lead to excessive antibody production. High proportions of Tfh cells have been associated with DSA formation in transplantation; therefore, Tfr cells are likely to play an important role in limiting DSA production. Understanding the signals that govern Tfr cell development and the balance between helper and regulatory function within the GC is key to understanding how these cells might be manipulated to reduce the risk of DSA development.This review discusses the development and function of Tfr cells and their relevance to transplantation. In particular how current and future immunosuppressive strategies might allow us to skew the ratio between Tfr and Tfh cells to increase or decrease the risk of de novo DSA formation.
Collapse
|
106
|
Abstract
Although cluster of differentiation (CD)8 regulatory T (Treg) cells have been in the last 20 years more studied since evidences of their role in tolerance as been demonstrated in transplantation, autoimmune diseases and cancer, their characteristics are still controversial. In this review, we will focus on recent advances on CD8 Treg cells and description of a role for CD8 Treg cells in tolerance in both solid organ transplantation and graft-versus-host disease and their potential for clinical trials.
Collapse
|
107
|
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: 50] [Impact Index Per Article: 8.3] [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
|
108
|
Ten Brinke A, Martinez-Llordella M, Cools N, Hilkens CMU, van Ham SM, Sawitzki B, Geissler EK, Lombardi G, Trzonkowski P, Martinez-Caceres E. Ways Forward for Tolerance-Inducing Cellular Therapies- an AFACTT Perspective. Front Immunol 2019; 10:181. [PMID: 30853957 PMCID: PMC6395407 DOI: 10.3389/fimmu.2019.00181] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
Clinical studies with cellular therapies using tolerance-inducing cells, such as tolerogenic antigen-presenting cells (tolAPC) and regulatory T cells (Treg) for the prevention of transplant rejection and the treatment of autoimmune diseases have been expanding the last decade. In this perspective, we will summarize the current perspectives of the clinical application of both tolAPC and Treg, and will address future directions and the importance of immunomonitoring in clinical studies that will result in progress in the field.
Collapse
Affiliation(s)
- Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marc Martinez-Llordella
- Department of Inflammation Biology, MRC Centre for Transplantation, School of Immunology and Microbial Sciences, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Catharien M U Hilkens
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Birgit Sawitzki
- Charité-Universitaetsmedizin Berlin, Berlin Institute of Health, Institute for Medical Immunology, Humboldt-Universitaet zu Berlin, Berlin, Germany
| | - Edward K Geissler
- Section of Experimental Surgery, Department of Surgery, University Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Giovanna Lombardi
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom
| | - Piotr Trzonkowski
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Eva Martinez-Caceres
- Division of Immunology, Germans Trias i Pujol University Hospital, LCMN, IGTP, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
109
|
Abstract
PURPOSE OF REVIEW The application of regulatory T cell (Treg) therapy in organ transplantation is actively being pursued using unmodified, typically polyclonal cells. As the results of these ongoing clinical trials emerge, it is time to plan the next wave of clinical trials of Tregs. Here we will review a key strategy to improve Treg effectiveness and reduce side effects, namely increasing Treg specificity - both in terms of antigen recognition and localization to the allograft. RECENT FINDINGS Study of chemokine signatures accompanying acute rejection has revealed several chemokines that could be targeted to increase Treg homing. For example, Tregs possessing a Th1-like phenotype and expressing CXCR3 are better able to migrate towards local inflammation. Allografts themselves can be modified to increase Treg-attracting chemokines and Tregs themselves can produce chemokines, facilitating local proximity to their targets of suppression. Finally, tailoring Treg antigen specificity by T-cell or chimeric antigen receptor engineering is another approach to increase the specificity of suppression and optimize localization. SUMMARY Treg localization to the graft is important, but the important role of lymph node and germinal center homing cannot be overlooked. There is an opportunity to learn from advances made in cancer immunotherapy to optimize Treg therapy for transplantation.
Collapse
|
110
|
Transient increase of activated regulatory T cells early after kidney transplantation. Sci Rep 2019; 9:1021. [PMID: 30705299 PMCID: PMC6355855 DOI: 10.1038/s41598-018-37218-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Regulatory T cells (Tregs) are crucial in controlling allospecific immune responses. However, studies in human kidney recipients regarding the contribution of polyspecific Tregs have provided differing results and studies on alloreactive Tregs are missing completely. In this retrospective study, we specifically analyzed activated CD4+CD25highFOXP3+GARP+ Tregs in 17 patients of a living donor kidney transplantation cohort longitudinally over 24 months by flow cytometry (FOXP3: forkhead box protein 3, GARP: glycoprotein A repetitions predominant). We could demonstrate that Tregs of patients with end-stage renal disease (ESRD) are already pre-activated when compared to healthy controls. Furthermore, even though total CD4+CD25highFOXP3+ Treg numbers decreased in the first three months after transplantation, frequency of activated Tregs increased significantly representing up to 40% of all peripheral Tregs. In a cohort of living donor kidney transplantation recipients with stable graft function, frequencies of activated Tregs did not correlate with the occurrence of acute cellular rejection or chronic graft dysfunction. Our results will be important for clinical trials using adoptive Treg therapy after kidney transplantation. Adoptively transferred Tregs could be important to compensate the Treg loss at month 3, while they have to compete within the Treg niche with a large number of activated Tregs.
Collapse
|
111
|
Romano M, Fanelli G, Albany CJ, Giganti G, Lombardi G. Past, Present, and Future of Regulatory T Cell Therapy in Transplantation and Autoimmunity. Front Immunol 2019; 10:43. [PMID: 30804926 PMCID: PMC6371029 DOI: 10.3389/fimmu.2019.00043] [Citation(s) in RCA: 377] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs) are important for the induction and maintenance of peripheral tolerance therefore, they are key in preventing excessive immune responses and autoimmunity. In the last decades, several reports have been focussed on understanding the biology of Tregs and their mechanisms of action. Preclinical studies have demonstrated the ability of Tregs to delay/prevent graft rejection and to control autoimmune responses following adoptive transfer in vivo. Due to these promising results, Tregs have been extensively studied as a potential new tool for the prevention of graft rejection and/or the treatment of autoimmune diseases. Currently, solid organ transplantation remains the treatment of choice for end-stage organ failure. However, chronic rejection and the ensuing side effects of immunosuppressants represent the main limiting factors for organ acceptance and patient survival. Autoimmune disorders are chronic diseases caused by the breakdown of tolerance against self-antigens. This is triggered either by a numerical or functional Treg defect, or by the resistance of effector T cells to suppression. In this scenario, patients receiving high doses of immunosuppressant are left susceptible to life-threatening opportunistic infections and have increased risk of malignancies. In the last 10 years, a few phase I clinical trials aiming to investigate safety and feasibility of Treg-based therapy have been completed and published, whilst an increasing numbers of trials are still ongoing. The first results showed safety and feasibility of Treg therapy and phase II clinical trials are already enrolling. In this review, we describe our understanding of Tregs focussing primarily on their ontogenesis, mechanisms of action and methods used in the clinic for isolation and expansion. Furthermore, we will describe the ongoing studies and the results from the first clinical trials with Tregs in the setting of solid organ transplantation and autoimmune disorders. Finally, we will discuss strategies to further improve the success of Treg therapy.
Collapse
Affiliation(s)
- Marco Romano
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giorgia Fanelli
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Caraugh Jane Albany
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giulio Giganti
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Scuola di Specializzazione in Medicina Interna, Universita' degli Studi di Milano, Milan, Italy
| | - Giovanna Lombardi
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
112
|
LeGuern C, Germana S. On the elusive TCR specificity of thymic regulatory T cells. Am J Transplant 2019; 19:15-20. [PMID: 30378738 DOI: 10.1111/ajt.15165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 01/25/2023]
Abstract
Therapies using thymus-derived regulatory T cells (Tregs) are promising strategies for preventing autoimmunity or graft rejection. The efficacy of these approaches is, however, contingent on a better understanding of Treg mode of action, especially about factors controlling their activation in vivo. Although key parameters of Treg suppression have been identified, little information is available on Treg activation in vivo via the TCR. In light of recent studies using TCR transgenic mouse models as well as unpublished data, we discuss evidence in support of the view that Treg TCR specificities are not necessarily highly diverse, that the accessibility of Treg selective antigens control Treg development, and that peptides derived from MHC class II (MHC-II) could be prevailing antigens involved in Treg selection. This novel perspective provides insights on Treg development as well as a conceptual basis to a significant contribution of MHC-II derived peptides in the shaping of the Treg TCR repertoire.
Collapse
Affiliation(s)
- Christian LeGuern
- Massachusetts General Hospital/Harvard Medical School - Center for Transplantation Sciences, Charlestown, Massachusetts
| | - Sharon Germana
- Massachusetts General Hospital/Harvard Medical School - Center for Transplantation Sciences, Charlestown, Massachusetts
| |
Collapse
|
113
|
Affiliation(s)
- Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, CA 94143, USA. .,Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
114
|
Regulatory T-cell therapy for autoimmune and autoinflammatory diseases: The next frontier. J Allergy Clin Immunol 2018; 142:1710-1718. [DOI: 10.1016/j.jaci.2018.10.015] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/12/2018] [Accepted: 10/19/2018] [Indexed: 02/08/2023]
|
115
|
Savage TM, Shonts BA, Obradovic A, Dewolf S, Lau S, Zuber J, Simpson MT, Berglund E, Fu J, Yang S, Ho SH, Tang Q, Turka LA, Shen Y, Sykes M. Early expansion of donor-specific Tregs in tolerant kidney transplant recipients. JCI Insight 2018; 3:124086. [PMID: 30429370 DOI: 10.1172/jci.insight.124086] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/10/2018] [Indexed: 12/23/2022] Open
Abstract
Allograft tolerance, in which a graft is accepted without long-term immunosuppression, could overcome numerous obstacles in transplantation. Human allograft tolerance has been intentionally induced across HLA barriers via combined kidney and bone marrow transplantation (CKBMT) with a regimen that induces only transient chimerism. Tregs are enriched early after CKBMT. While deletional tolerance contributes to long-term tolerance, the role of Tregs remains unclear. We have optimized a method for identifying the donor-specific Treg repertoire and used it to interrogate the fate of donor-specific Tregs after CKBMT. We expanded Tregs with several different protocols. Using functional analyses and T cell receptor sequencing, we found that expanding sorted Tregs with activated donor B cells identified the broadest Treg repertoire with the greatest potency and donor specificity of suppression. This method outperformed both alloantigen stimulation with CTLA4Ig and sequencing of CFSElo cells from the primary mixed lymphocyte reaction. In 3 tolerant and 1 nontolerant CKBMT recipients, we sequenced donor-specific Tregs before transplant and tracked them after transplant. Preexisting donor-specific Tregs were expanded at 6 months after CKBMT in tolerant patients and were reduced in the nontolerant patient. These results suggest that early expansion of donor-specific Tregs is involved in tolerance induction following CKBMT.
Collapse
Affiliation(s)
- Thomas M Savage
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Brittany A Shonts
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Susan Dewolf
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Saiping Lau
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Julien Zuber
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Michael T Simpson
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Erik Berglund
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Suxiao Yang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Siu-Hong Ho
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Qizhi Tang
- Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Laurence A Turka
- Center for Translational Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA.,Immune Tolerance Network, Bethesda, Maryland, USA
| | - Yufeng Shen
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, New York, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, New York, USA.,Department of Microbiology & Immunology, Columbia University Medical Center, Columbia University, New York, New York, USA.,Department of Surgery, Columbia University Medical Center, Columbia University, New York, New York, USA
| |
Collapse
|
116
|
Yang ZG, Wen RT, Feng JS, Cao PJ, Zhou HT, Liu WX. Recipient-Derived Allo-iTregs Induced by Donor DCs Effectively Inhibit the Proliferation of Donor T Cells and Reduce GVHD. Anat Rec (Hoboken) 2018; 302:825-836. [PMID: 30312018 DOI: 10.1002/ar.23972] [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: 04/15/2018] [Revised: 05/10/2018] [Accepted: 06/04/2018] [Indexed: 11/09/2022]
Abstract
To compare the potency of recipient-derived, antigen-specific regulatory T cells induced by different dendritic cells (DCs; iTregs) and freshly isolated natural regulatory T cells (nTregs) in preventing mouse graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (BMT). CD4+ T cells from recipient BALB/c mice were stimulated with DCs from recipient BALB/c (syn-DCs), donor B6 (allo-DCs), and third-party C3H (third-party-DCs) mice to induce different iTregs. In parallel, nTregs were isolated from spleen cells of recipient BALB/c (syn-nTregs) and donor B6 (allo-nTregs) mice using magnetic-activated cell sorting. Mixed lymphocyte reaction (MLR) assays were performed to evaluate the suppressive ability of these various regulatory T cells (Tregs). Both the iTregs and nTregs were transfused to GVHD mice on Days 0, 1, 3, and 5. Body weight, GVHD score, and survival time were monitored. Peripheral Tregs were subsequently examined on Days 7, 14, 21, and 28 after BMT, while chimerism was evaluated on Days 14 and 60. Histopathology of colon, liver, and spleen were also performed. DCs markedly induced CD25+ and Foxp3+ expression on CD4+ T cells. The allo-DC-induced Tregs (allo-iTregs) suppressed the proliferation of alloreactive T cells better than the other iTregs/nTregs in MLR assays (P < 0.05). Meanwhile, transfusion of the allo-iTregs reduced the severity of GVHD (P < 0.05), increased survival time compared with the GVHD group (P < 0.05), and enhanced the chimerism proportion. On Day 28 after BMT, the allo-iTregs group had the highest frequency of peripheral Tregs (P < 0.05). Recipient-derived allo-iTregs induced by donor DCs included predominant clones that specifically recognized donor antigens. These allo-iTregs not only prevented GVHD by suppressing the proliferation of donor-alloreactive T cells, but also promoted engraftment, and prolonged the survival of GVHD mice. Anat Rec, 2018. © 2018 Wiley Periodicals, Inc. Anat Rec, 302:825-836, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zhi-Gang Yang
- Affiliated Central People's Hospital of Zhanjiang of Guangdong Medical University, Zhanjiang, Guangdong, 524045, People's Republic of China.,Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Rui-Ting Wen
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Jin-Shan Feng
- Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Pei-Jie Cao
- Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Hai-Tao Zhou
- Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, 510000, People's Republic of China
| | - Wen-Xin Liu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| |
Collapse
|
117
|
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: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
118
|
Watanabe M, Kumagai-Braesch M, Yao M, Thunberg S, Berglund D, Sellberg F, Jorns C, Enoksson SL, Henriksson J, Lundgren T, Uhlin M, Berglund E, Ericzon BG. Ex Vivo Generation of Donor Antigen-Specific Immunomodulatory Cells: A Comparison Study of Anti-CD80/86 mAbs and CTLA4-lg Costimulatory Blockade. Cell Transplant 2018; 27:1692-1704. [PMID: 30261751 PMCID: PMC6299197 DOI: 10.1177/0963689718794642] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Adoptive transfer of alloantigen-specific immunomodulatory cells generated ex vivo with anti-CD80/CD86 mAbs (2D10.4/IT2.2) holds promise for operational tolerance after transplantation. However, good manufacturing practice is required to allow widespread clinical application. Belatacept, a clinically approved cytotoxic T-lymphocyte antigen 4-immunoglobulin that also binds CD80/CD86, could be an alternative agent for 2D10.4/IT2.2. With the goal of generating an optimal cell treatment with clinically approved reagents, we evaluated the donor-specific immunomodulatory effects of belatacept- and 2D10.4/IT2.2-generated immunomodulatory cells. Immunomodulatory cells were generated by coculturing responder human peripheral blood mononuclear cells (PBMCs) (50 × 106 cells) with irradiated donor PBMCs (20 × 106 cells) from eight human leukocyte antigen-mismatched responder–donor pairs in the presence of either 2D10.4/IT2.2 (3 μg/106 cells) or belatacept (40 μg/106 cells). After 14 days of coculture, the frequencies of CD4+ T cells, CD8+ T cells, and natural killer cells as well as interferon gamma (IFN-γ) production in the 2D10.4/IT2.2- and belatacept-treated groups were lower than those in the control group. The percentage of CD19+ B cells was higher in the 2D10.4/IT2.2- and belatacept-treated groups than in the control group. The frequency of CD4+CD25+CD127lowFOXP3+ T cells increased from 4.1±1.0% (preculture) to 7.1±2.6% and 7.3±2.6% (day 14) in the 2D10.4/IT2.2- and belatacept-treated groups, respectively (p<0.05). Concurrently, delta-2 FOXP3 mRNA expression increased significantly. Compared with cells derived from the no-antibody treated control group, cells generated from both the 2D10.4/IT2.2- and belatacept-treated groups produced lower IFN-γ and higher interleukin-10 levels in response to donor-antigens, as detected by enzyme-linked immunospot. Most importantly, 2D10.4/IT2.2- and belatacept-generated cells effectively impeded the proliferative responses of freshly isolated responder PBMCs against donor-antigens. Our results indicate that belatacept-generated donor-specific immunomodulatory cells possess comparable phenotypes and immunomodulatory efficacies to those generated with 2D10.4/IT2.2. We suggest that belatacept could be used for ex vivo generation of clinical grade alloantigen-specific immunomodulatory cells for tolerance induction after transplantation.
Collapse
Affiliation(s)
- M Watanabe
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - Makiko Kumagai-Braesch
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - M Yao
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - S Thunberg
- Department of Clinical Immunology, Karolinska University Hospital, Stockholm, Sweden
| | - D Berglund
- Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
| | - F Sellberg
- Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Uppsala, Sweden
| | - C Jorns
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - S Lind Enoksson
- Department of Clinical Immunology, Karolinska University Hospital, Stockholm, Sweden
| | - J Henriksson
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - T Lundgren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - M Uhlin
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Immunology, Karolinska University Hospital, Stockholm, Sweden
| | - E Berglund
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| | - B-G Ericzon
- Division of Transplantation Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Department of Transplantation Surgery, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
119
|
Abstract
PURPOSE OF REVIEW The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine's extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|
120
|
Romano M, Fanelli G, Tan N, Nova-Lamperti E, McGregor R, Lechler RI, Lombardi G, Scottà C. Expanded Regulatory T Cells Induce Alternatively Activated Monocytes With a Reduced Capacity to Expand T Helper-17 Cells. Front Immunol 2018; 9:1625. [PMID: 30079063 PMCID: PMC6062605 DOI: 10.3389/fimmu.2018.01625] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/02/2018] [Indexed: 12/29/2022] Open
Abstract
Regulatory T cells (Tregs) are essential in maintaining peripheral immunological tolerance by modulating several subsets of the immune system including monocytes. Under inflammatory conditions, monocytes migrate into the tissues, where they differentiate into dendritic cells or tissue-resident macrophages. As a result of their context-dependent plasticity, monocytes have been implicated in the development/progression of graft-vs-host disease (GvHD), autoimmune diseases and allograft rejection. In the last decade, Tregs have been exploited for their use in cell therapy with the aim to induce tolerance after solid organ transplantation and for the treatment of autoimmune diseases and GvHD. To date, safety and feasibility of Treg infusion has been demonstrated; however, many questions of how these cells induce tolerance have been raised and need to be answered. As monocytes constitute the major cellular component in inflamed tissues, we have developed an in vitro model to test how Tregs modulate their phenotype and function. We demonstrated that expanded Tregs can drive monocytes toward an alternatively activated state more efficiently than freshly isolated Tregs. The effect of expanded Tregs on monocytes led to a reduced production of pro-inflammatory cytokines (IL-6 and tumor necrosis factor-α) and NF-κB activation. Furthermore, monocytes co-cultured with expanded Tregs downregulated the expression of co-stimulatory and MHC-class II molecules with a concomitant upregulation of M2 macrophage specific markers, CD206, heme oxygenase-1, and increased interleukin-10 production. Importantly, monocytes co-cultured with expanded Tregs showed a reduced capacity to expand IL-17-producing T cells compared with monocyte cultured with freshly isolated Tregs and conventional T cells. The capacity to decrease the expansion of pro-inflammatory Th-17 was not cytokine mediated but the consequence of their lower expression of the co-stimulatory molecule CD86. Our data suggest that expanded Tregs have the capacity to induce phenotypical and functional changes in monocytes that might be crucial for tolerance induction in transplantation and the prevention/treatment of GvHD and autoimmune diseases.
Collapse
Affiliation(s)
- Marco Romano
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giorgia Fanelli
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Nicole Tan
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Estefania Nova-Lamperti
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Concepcion, Chile
| | - Reuben McGregor
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Robert I Lechler
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Cristiano Scottà
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
121
|
Landman S, de Oliveira VL, van Erp PEJ, Fasse E, Bauland SCG, Joosten I, Koenen HJPM. Intradermal injection of low dose human regulatory T cells inhibits skin inflammation in a humanized mouse model. Sci Rep 2018; 8:10044. [PMID: 29968819 PMCID: PMC6030170 DOI: 10.1038/s41598-018-28346-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 06/11/2018] [Indexed: 12/31/2022] Open
Abstract
Recent regulatory T cell (Treg) based clinical trials support their therapeutic potential in transplantation and auto-inflammatory diseases. However, large numbers of Treg are needed to accomplish therapeutic efficacy. Local injection at the site of inflammation (targeted delivery) may lower the numbers needed for therapy. We evaluated if local delivery of low numbers of human Treg by intradermal injection was able to prevent skin inflammation, using the humanized mouse huPBL-SCID-huSkin allograft model. A dose of only 1 × 105 freshly isolated, non expanded Treg injected intradermally in close proximity to the transplanted human skin prevented inflammation of the grafted tissue induced by 4 × 107 IP injected human allogeneic PBMCs, (ratio Treg:PBMC = 1:400), as indicated by the inhibition of epidermal thickening, sustained Keratin-10 expression, the absence of Keratin-16 up regulation and prevention of human CD3+ T cell influx. A concomitant reduction of human T cells was observed in lymph nodes and spleen of the mice. Injection of Treg at the contralateral side was also shown to inhibit skin inflammation, suggesting that the inflammatory response was regulated both locally and systemically. In conclusion, local application of Treg may be an attractive way to suppress inflammation in vivo without the need for prior ex vivo expansion.
Collapse
Affiliation(s)
- Sija Landman
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Vivian L de Oliveira
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Piet E J van Erp
- Radboud university medical center, department of Dermatology, Nijmegen, The Netherlands
| | - Esther Fasse
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | | | - Irma Joosten
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Radboud university medical center, department of Laboratory Medicine-Medical Immunology, Nijmegen, The Netherlands.
| |
Collapse
|
122
|
Whitehouse GP, Hope A, Sanchez-Fueyo A. Regulatory T-cell therapy in liver transplantation. Transpl Int 2018; 30:776-784. [PMID: 28608637 DOI: 10.1111/tri.12998] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/27/2017] [Accepted: 06/07/2017] [Indexed: 12/24/2022]
Abstract
Modern immunosuppression drug regimens have produced excellent short-term survival after liver transplantation but it is generally accepted that the side effects of these medications remain a significant contributing factor for less satisfactory long term outcomes. The liver has unique tolerogenic properties as evidenced by the higher rates of operational tolerance seen in liver transplant recipients compared to other solid organ transplants, and therefore, liver transplantation offers an attractive setting in which to study tolerizing therapies. CD4+ CD25+ FOXP3+ regulatory T cells (Tregs) are crucial for maintenance of self-tolerance and prevention of autoimmune disease and are therefore an appealing potential candidate for use as a tolerizing cell therapy. In this review, we summarize the evidence from drug withdrawal trials of spontaneous operational tolerance in liver transplantation, the unique immunology of the hepatic microenvironment, the evidence for the use of CD4+ CD25+ FOXP3+ regulatory T cells as a tolerance inducing therapy in liver transplantation and the challenges in producing clinical grade Treg cell products.
Collapse
Affiliation(s)
- Gavin P Whitehouse
- Division of Transplantation Immunology and Mucosal Biology, Institute of Liver Studies, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Andrew Hope
- CRF GMP Unit, NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Alberto Sanchez-Fueyo
- Division of Transplantation Immunology and Mucosal Biology, Institute of Liver Studies, Medical Research Council Centre for Transplantation, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
123
|
Kawai K, Uchiyama M, Hester J, Wood K, Issa F. Regulatory T cells for tolerance. Hum Immunol 2018; 79:294-303. [DOI: 10.1016/j.humimm.2017.12.013] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/16/2017] [Accepted: 12/26/2017] [Indexed: 12/29/2022]
|
124
|
|
125
|
Abstract
CD4+CD25highFoxP3+ T regulatory cells (Tregs) are immunodominant suppressors in the immune system. Tregs use various mechanisms to control immune responses. Preclinical data from animal models have confirmed the huge therapeutic potential of Tregs in many immune-mediated diseases. Hence, these cells are now on the road to translation to cell therapy in the clinic as the first clinical trials are accomplished. To date, clinical research has involved mainly hematopoietic stem cell transplantations, solid organ transplantations, and autoimmunity. Despite difficulties with legislation and technical issues, treatment is constantly evolving and may soon represent a valid alternative for patients with diseases that are currently incurable. This review focuses on the basic and clinical experience with Tregs with adoptive transfer of these cells, primarily from clinical trials, as well as on perspectives on clinical use and technical problems with implementing the therapy.
Collapse
|
126
|
Safinia N, Grageda N, Scottà C, Thirkell S, Fry LJ, Vaikunthanathan T, Lechler RI, Lombardi G. Cell Therapy in Organ Transplantation: Our Experience on the Clinical Translation of Regulatory T Cells. Front Immunol 2018. [PMID: 29535728 PMCID: PMC5834909 DOI: 10.3389/fimmu.2018.00354] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Solid organ transplantation is the treatment of choice for patients with end-stage organ dysfunction. Despite improvements in short-term outcome, long-term outcome is suboptimal due to the increased morbidity and mortality associated with the toxicity of immunosuppressive regimens and chronic rejection (1–5). As such, the attention of the transplant community has focused on the development of novel therapeutic strategies to achieve allograft tolerance, a state whereby the immune system of the recipient can be re-educated to accept the allograft, averting the need for long-term immunosuppression. Indeed, reports of “operational” tolerance, whereby the recipient is off all immunosuppressive drugs and maintaining good graft function, is well documented in the literature for both liver and kidney transplantations (6–8). However, this phenomenon is rare and in the setting of liver transplantation has been shown to occur late after transplantation, with the majority of patients maintained on life-long immunosupression to prevent allograft rejection (9). As such, significant research has focused on immune regulation in the context of organ transplantation with regulatory T cells (Tregs) identified as cells holding considerable promise in this endeavor. This review will provide a brief introduction to human Tregs, their phenotypic and functional characterization and focuses on our experience to date at the clinical translation of Treg immunotherapy in the setting of solid organ transplantation.
Collapse
Affiliation(s)
- Niloufar Safinia
- Department of Immunoregulation and Immune Intervention, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom.,Faculty of Medicine, Division of Digestive Disease, Imperial College London, London, United Kingdom
| | - Nathali Grageda
- Department of Immunoregulation and Immune Intervention, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Cristiano Scottà
- Department of Immunoregulation and Immune Intervention, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Sarah Thirkell
- Department of Immunoregulation and Immune Intervention, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Laura J Fry
- Clinical Research Facility GMP Unit, NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Trishan Vaikunthanathan
- The Blizard Institute of Cell and Molecular Science, Queen Mary University of London, London, United Kingdom
| | - Robert I Lechler
- Department of Immunoregulation and Immune Intervention, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Department of Immunoregulation and Immune Intervention, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
127
|
Duggleby R, Danby RD, Madrigal JA, Saudemont A. Clinical Grade Regulatory CD4 + T Cells (Tregs): Moving Toward Cellular-Based Immunomodulatory Therapies. Front Immunol 2018; 9:252. [PMID: 29487602 PMCID: PMC5816789 DOI: 10.3389/fimmu.2018.00252] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/29/2018] [Indexed: 12/26/2022] Open
Abstract
Regulatory T cells (Tregs) are CD4+ T cells that are key players of immune tolerance. They are powerful suppressor cells, able to impact the function of numerous immune cells, including key effectors of inflammation such as effector T cells. For this reason, Tregs are an ideal candidate for the development of cell therapy approaches to modulate immune responses. Treg therapy has shown promising results so far, providing key knowledge on the conditions in which these cells can provide protection and demonstrating that they could be an alternative to current pharmacological immunosuppressive therapies. However, a more comprehensive understanding of their characteristics, isolation, activation, and expansion is needed to be able design cost effective therapies. Here, we review the practicalities of making Tregs a viable cell therapy, in particular, discussing the challenges faced in isolating and manufacturing Tregs and defining what are the most appropriate applications for this new therapy.
Collapse
Affiliation(s)
- Richard Duggleby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Robert David Danby
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom.,Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| | - Aurore Saudemont
- Anthony Nolan Research Institute, London, United Kingdom.,University College London, London, United Kingdom
| |
Collapse
|
128
|
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: 53] [Impact Index Per Article: 7.6] [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
|
129
|
Fraser H, Safinia N, Grageda N, Thirkell S, Lowe K, Fry LJ, Scottá C, Hope A, Fisher C, Hilton R, Game D, Harden P, Bushell A, Wood K, Lechler RI, Lombardi G. A Rapamycin-Based GMP-Compatible Process for the Isolation and Expansion of Regulatory T Cells for Clinical Trials. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 8:198-209. [PMID: 29552576 PMCID: PMC5850906 DOI: 10.1016/j.omtm.2018.01.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/16/2018] [Indexed: 01/09/2023]
Abstract
The concept of regulatory T cell (Treg)-based immunotherapy has enormous potential for facilitating tolerance in autoimmunity and transplantation. Clinical translation of Treg cell therapy requires production processes that satisfy the rigors of Good Manufacturing Practice (GMP) standards. In this regard, we report our findings on the implementation of a robust GMP compliant process for the ex vivo expansion of clinical grade Tregs, demonstrating the feasibility of this developed process for the manufacture of a final product for clinical application. This Treg isolation procedure ensured the selection of a pure Treg population that underwent a 300-fold expansion after 36 days of culture, while maintaining a purity of more than 75% CD4+CD25+FOXP3+ cells and a suppressive function of above 80%. Furthermore, we report the successful cryopreservation of the final product, demonstrating the maintenance of phenotype and function. The process outlined in this manuscript has been implemented in the ONE study, a multicenter phase I/IIa clinical trial in which cellular therapy is investigated in renal transplantation.
Collapse
Affiliation(s)
- Henrieta Fraser
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Niloufar Safinia
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Nathali Grageda
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Sarah Thirkell
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Katie Lowe
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Laura J Fry
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Cristiano Scottá
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Andrew Hope
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Christopher Fisher
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Rachel Hilton
- The Department of Nephrology and Transplantation, Guy's Hospital, Guy's and St. Thomas NHS Foundation Trust
| | - David Game
- The Department of Nephrology and Transplantation, Guy's Hospital, Guy's and St. Thomas NHS Foundation Trust
| | | | - Andrew Bushell
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Kathryn Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Robert I Lechler
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| | - Giovanna Lombardi
- Division of Transplantation, Immunology and Mucosal Biology, King's College London, London, UK
| |
Collapse
|
130
|
Mathew JM, Ansari MJ, Gallon L, Leventhal JR. Cellular and functional biomarkers of clinical transplant tolerance. Hum Immunol 2018; 79:322-333. [PMID: 29374560 DOI: 10.1016/j.humimm.2018.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/16/2022]
Abstract
Development of tolerance protocols requires assays or biomarkers that distinguish tolerant recipients from non-tolerant ones to be established. In addition, a thorough understanding of the plausible mechanisms associated with clinical transplant tolerance is necessary to take the field forward. Unlike the majority of molecular signature analyses utilized by others, the emphasis of this article is on the cellular and functional biomarkers of induced transplant tolerance. Immunity to an organ transplant is very complex, comprised of two broad categories - innate and acquired or adaptive immune responses. Innate immunity can be avoided by eliminating or preventing ischemic injuries to the donor organ and tolerance at the level of adaptive immunity can be induced by infusions of a number of cellular products. Since adaptive immune response consists of inflammatory hypersensitivity, cellular (cytotoxic and helper) and humoral aspects, all these need to be measured, and the recipients who demonstrate donor-specific unresponsiveness in all can be considered tolerant or candidates for immunosuppression minimization and/or withdrawal. The mechanisms by which these agents bring about transplant tolerance include regulation, anergy, exhaustion, senescence and deletion of the recipient immune cells. Another proven mechanism of tolerance is full or mixed donor chimerism. However, it should be cautioned that non-deletional tolerance can be reversed.
Collapse
Affiliation(s)
- James M Mathew
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA; Department of Microbiology-Immunology, Northwestern University, Chicago, IL, USA.
| | - Mohammed Javeed Ansari
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA; Department of Medicine-Nephrology, Northwestern University, Chicago, IL, USA
| | - Lorenzo Gallon
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA; Department of Medicine-Nephrology, Northwestern University, Chicago, IL, USA
| | - Joseph R Leventhal
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA
| |
Collapse
|
131
|
Generation and Characterization of Alloantigen-Specific Regulatory T Cells For Clinical Transplant Tolerance. Sci Rep 2018; 8:1136. [PMID: 29348660 PMCID: PMC5773708 DOI: 10.1038/s41598-018-19621-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/05/2018] [Indexed: 01/28/2023] Open
Abstract
Donor-specific CD4+CD127−CD25+FOXP3+ regulatory T cells (AgTregs) have the potential to induce clinical transplant tolerance; however, their expansion ex vivo remains challenging. We optimized a novel expansion protocol to stimulate donor-specific Tregs using soluble 4-trimer CD40 ligand (CD40L)-activated donor B cells that expressed mature antigen-presenting cell markers. This avoided the use of CD40L-expressing stimulator cells that might otherwise result in potential cellular contamination. Purified allogeneic “recipient” CD4+CD25+ Tregs were stimulated on days 0 and 7 with expanded “donor” B cells in the presence of IL-2, TGFβ and sirolimus (SRL). Tregs were further amplified by polyclonal stimulation with anti-CD3/CD28 beads on day 14 without SRL, and harvested on day 21, with extrapolated fold expansion into the thousands. The expanded AgTregs maintained expression of classical Treg markers including demethylation of the Treg-specific demethylated region (CNS2) and also displayed constricted TcR repertoire. We observed AgTregs more potently inhibited MLR than polyclonally expanded Tregs and generated new Tregs in autologous responder cells (a measure of infectious tolerance). Thus, an optimized and more clinically applicable protocol for the expansion of donor-specific Tregs has been developed.
Collapse
|
132
|
Fuchs A, Gliwiński M, Grageda N, Spiering R, Abbas AK, Appel S, Bacchetta R, Battaglia M, Berglund D, Blazar B, Bluestone JA, Bornhäuser M, Ten Brinke A, Brusko TM, Cools N, Cuturi MC, Geissler E, Giannoukakis N, Gołab K, Hafler DA, van Ham SM, Hester J, Hippen K, Di Ianni M, Ilic N, Isaacs J, Issa F, Iwaszkiewicz-Grześ D, Jaeckel E, Joosten I, Klatzmann D, Koenen H, van Kooten C, Korsgren O, Kretschmer K, Levings M, Marek-Trzonkowska NM, Martinez-Llordella M, Miljkovic D, Mills KHG, Miranda JP, Piccirillo CA, Putnam AL, Ritter T, Roncarolo MG, Sakaguchi S, Sánchez-Ramón S, Sawitzki B, Sofronic-Milosavljevic L, Sykes M, Tang Q, Vives-Pi M, Waldmann H, Witkowski P, Wood KJ, Gregori S, Hilkens CMU, Lombardi G, Lord P, Martinez-Caceres EM, Trzonkowski P. Minimum Information about T Regulatory Cells: A Step toward Reproducibility and Standardization. Front Immunol 2018; 8:1844. [PMID: 29379498 PMCID: PMC5775516 DOI: 10.3389/fimmu.2017.01844] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 12/06/2017] [Indexed: 12/13/2022] Open
Abstract
Cellular therapies with CD4+ T regulatory cells (Tregs) hold promise of efficacious treatment for the variety of autoimmune and allergic diseases as well as posttransplant complications. Nevertheless, current manufacturing of Tregs as a cellular medicinal product varies between different laboratories, which in turn hampers precise comparisons of the results between the studies performed. While the number of clinical trials testing Tregs is already substantial, it seems to be crucial to provide some standardized characteristics of Treg products in order to minimize the problem. We have previously developed reporting guidelines called minimum information about tolerogenic antigen-presenting cells, which allows the comparison between different preparations of tolerance-inducing antigen-presenting cells. Having this experience, here we describe another minimum information about Tregs (MITREG). It is important to note that MITREG does not dictate how investigators should generate or characterize Tregs, but it does require investigators to report their Treg data in a consistent and transparent manner. We hope this will, therefore, be a useful tool facilitating standardized reporting on the manufacturing of Tregs, either for research purposes or for clinical application. This way MITREG might also be an important step toward more standardized and reproducible testing of the Tregs preparations in clinical applications.
Collapse
Affiliation(s)
- Anke Fuchs
- GMP facility, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), and Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mateusz Gliwiński
- Department of Clinical Immunology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | - Nathali Grageda
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, United Kingdom
| | - Rachel Spiering
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Abul K Abbas
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
| | - Silke Appel
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rosa Bacchetta
- Pediatric Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
| | - Manuela Battaglia
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, and TrialNet Clinical Center, San Raffaele Hospital, Milan, Italy
| | - David Berglund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bruce Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minnesota, MN, United States
| | - Jeffrey A Bluestone
- Hormone Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Martin Bornhäuser
- GMP facility, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), and Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Academic Medical Center, Amsterdam, Netherlands
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL, United States
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Maria Cristina Cuturi
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France
| | - Edward Geissler
- Division of Experimental Surgery, Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Nick Giannoukakis
- Allegheny Health Network, Institute of Cellular Therapeutics, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Karolina Gołab
- Transplant Institute, Department of Surgery, The University of Chicago, Chicago, IL, United States
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of Amsterdam, Academic Medical Center, Amsterdam, Netherlands
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Keli Hippen
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minnesota, MN, United States
| | - Mauro Di Ianni
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Natasa Ilic
- Department for Immunology and Immunoparasitology, National Reference Laboratory for Trichinellosis, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - John Isaacs
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,National Institute for Health Research Newcastle Biomedical Research Centre at Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Endocrinology, Diabetology, Transplantationsforschungszentrum, Medical School of Hannover (MHH), Hannover, Germany
| | - Irma Joosten
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, Netherlands
| | - David Klatzmann
- Immunology-Immunopathology-Immunotherapy (i3), UPMC Univ Paris 06, UMRS 959, Sorbonne Université, and Biotherapy (CIC-BTi) and Inflammation-Immunopathology-Biotherapy Department, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hans Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, Netherlands
| | - Cees van Kooten
- Department of Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University Hospital, Uppsala, Sweden.,Transplantation Immunology, Gothenburg University, Gothenburg, Sweden
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, and Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Hospital and Medical Faculty Carl Gustav Carus of TU Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Megan Levings
- Department of Surgery, Faculty of Medicine, The University of British Columbia, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Natalia Maria Marek-Trzonkowska
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Marc Martinez-Llordella
- Medical Research Council Centre for Transplantation, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Djordje Miljkovic
- Department of Immunology, IBISS, University of Belgrade, Belgrade, Serbia
| | - Kingston H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Joana P Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Ciriaco A Piccirillo
- Departments of Microbiology & Immunology and Medicine, Faculty of Medicine, McGill University, Program in Infectious Disease and Immunity in Global Health, Centre of Excellence in Translational Immunology (CETI), Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Amy L Putnam
- Hormone Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Thomas Ritter
- College of Medicine, Nursing and Health Sciences, Regenerative Medicine Institute (REMEDI), Biomedical Sciences, National University of Ireland, Galway, Ireland
| | - Maria Grazia Roncarolo
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, ISCBRM, Stanford School of Medicine, Stanford, CA, United States
| | - Shimon Sakaguchi
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Silvia Sánchez-Ramón
- Department of Clinical Immunology, Hospital Clínico San Carlos, Universidad Complutense of Madrid, Madrid, Spain
| | - Birgit Sawitzki
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Ljiljana Sofronic-Milosavljevic
- Department for Immunology and Immunoparasitology, National Reference Laboratory for Trichinellosis, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Megan Sykes
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, Bone Marrow Transplantation Research, Division of Hematology/Oncology, Columbia University Medical Center, Columbia University, New York, NY, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Marta Vives-Pi
- Immunology of Diabetes Unit, Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Herman Waldmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Piotr Witkowski
- Transplant Institute, Department of Surgery, The University of Chicago, Chicago, IL, United States
| | - Kathryn J Wood
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Group, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Catharien M U Hilkens
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Giovanna Lombardi
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, United Kingdom
| | - Phillip Lord
- School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Eva M Martinez-Caceres
- Immunology Division, Germans Trias i Pujol University Hospital - Can Ruti, Department Cellular Biology, Physiology, Immunology, Universitat Autònoma Barcelona, Badalona, Spain
| | - Piotr Trzonkowski
- Department of Clinical Immunology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
133
|
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: 10] [Impact Index Per Article: 1.4] [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
|
134
|
Kang K, Chung J, Yang J, Kim H. Current Perspectives on Emerging CAR-Treg Cell Therapy: Based on Treg Cell Therapy in Clinical Trials and the Recent Approval of CAR-T Cell Therapy. KOREAN JOURNAL OF TRANSPLANTATION 2017. [DOI: 10.4285/jkstn.2017.31.4.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Koeun Kang
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jaeseok Yang
- Transplantation Center, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyori Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| |
Collapse
|
135
|
Anti-donor regulatory T cell therapy in liver transplantation. Hum Immunol 2017; 79:288-293. [PMID: 29292027 DOI: 10.1016/j.humimm.2017.12.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/14/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022]
Abstract
Liver transplantation is accepted as the most reliable therapeutic option for patients with end-stage liver failure, but lifelong administration of immunosuppressive agents continues to be problematic due to various drug-induced morbidities and the risk of mortality. Complete cessation of immunosuppressive drugs while maintaining normal graft function and histology, called operational tolerance, has the potential to overcome these long-standing problems. Previously, we reported the results of a pilot study of anti- donor regulatory T cell therapy in 10 consecutive adult patients who underwent living donor liver transplantation (LDLT), of whom 7 patients successfully stopped immunosuppression for nearly 2 years. Described herein are the clinical follow-ups of these patients, a brief description of the protocol and its theoretical background, and a possible explanation for the immunological findings.
Collapse
|
136
|
Scott DW. From IgG Fusion Proteins to Engineered-Specific Human Regulatory T Cells: A Life of Tolerance. Front Immunol 2017; 8:1576. [PMID: 29181011 PMCID: PMC5693857 DOI: 10.3389/fimmu.2017.01576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/02/2017] [Indexed: 01/23/2023] Open
Abstract
Recent efforts have concentrated on approaches to expand and “specify” human regulatory T cells (Tregs) and to apply them to modulate adverse immune responses in autoimmunity and hemophilia. We have used retroviral transduction of specific T-cell receptor, single chain Fv, or antigen domains in Tregs to achieve this goal. Each of these approaches have advantages and disadvantages. Results with these engineered T cells and evolution of the research developments and paths that led to the development of specific regulatory approaches for tolerance are summarized.
Collapse
Affiliation(s)
- David W Scott
- Department of Medicine, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
137
|
Yang JH, Eun SC. Therapeutic application of T regulatory cells in composite tissue allotransplantation. J Transl Med 2017; 15:218. [PMID: 29073905 PMCID: PMC5658973 DOI: 10.1186/s12967-017-1322-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
With growing number of cases in recent years, composite tissue allotransplantation (CTA) has been improving the quality of life of patient who seeks reconstruction and repair of damaged tissues. Composite tissue allografts are heterogeneous. They are composed of a variety of tissue types, including skin, muscle, vessel, bone, bone marrow, lymph nodes, nerve, and tendon. As a primary target of CTA, skin has high antigenicity with a rich repertoire of resident cells that play pivotal roles in immune surveillance. In this regard, understanding the molecular mechanisms involved in immune rejection in the skin would be essential to achieve successful CTA. Although scientific evidence has proved the necessity of immunosuppressive drugs to prevent rejection of allotransplanted tissues, there remains a lingering dilemma due to the lack of specificity of targeted immunosuppression and risks of side effects. A cumulative body of evidence has demonstrated T regulatory (Treg) cells have critical roles in induction of immune tolerance and immune homeostasis in preclinical and clinical studies. Presently, controlling immune susceptible characteristics of CTA with adoptive transfer of Treg cells is being considered promising and it has drawn great interests. This updated review will focus on a dominant form of Treg cells expressing CD4+CD25+ surface molecules and a forkhead box P3 transcription factor with immune tolerant and immune homeostasis activities. For future application of Treg cells as therapeutics in CTA, molecular and cellular characteristics of CTA and immune rejection, Treg cell development and phenotypes, Treg cell plasticity and stability, immune tolerant functions of Treg cells in CTA in preclinical studies, and protocols for therapeutic application of Treg cells in clinical settings are addressed in this review. Collectively, Treg cell therapy in CTA seems feasible with promising perspectives. However, the extreme high immunogenicity of CTA warrants caution.
Collapse
Affiliation(s)
- Jeong-Hee Yang
- Department of Plastic and Reconstructive Surgery, Composite Tissue Allotransplantation Immunology Laboratory, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seok-Chan Eun
- Department of Plastic and Reconstructive Surgery, Composite Tissue Allotransplantation Immunology Laboratory, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
138
|
Harnessing Advances in T Regulatory Cell Biology for Cellular Therapy in Transplantation. Transplantation 2017; 101:2277-2287. [PMID: 28376037 DOI: 10.1097/tp.0000000000001757] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular therapy with CD4FOXP3 T regulatory (Treg) cells is a promising strategy to induce tolerance after solid-organ transplantation or prevent graft-versus-host disease after transfer of hematopoietic stem cells. Treg cells currently used in clinical trials are either polyclonal, donor- or antigen-specific. Aside from variations in isolation and expansion protocols, however, most therapeutic Treg cell-based products are much alike. Ongoing basic science work has provided considerable new insight into multiple facets of Treg cell biology, including their stability, homing, and functional specialization; integrating these basic science discoveries with clinical efforts will support the development of next-generation therapeutic Treg cells with enhanced efficacy. In this review, we summarize recent advances in knowledge of how Treg cells home to lymphoid and peripheral tissues, and control antibody production and tissue repair. We also discuss newly appreciated pathways that modulate context-specific Treg cell function and stability. Strategies to improve and tailor Treg cells for cell therapy to induce transplantation tolerance are highlighted.
Collapse
|
139
|
Passerini L, Bacchetta R. Forkhead-Box-P3 Gene Transfer in Human CD4 + T Conventional Cells for the Generation of Stable and Efficient Regulatory T Cells, Suitable for Immune Modulatory Therapy. Front Immunol 2017; 8:1282. [PMID: 29075264 PMCID: PMC5643480 DOI: 10.3389/fimmu.2017.01282] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022] Open
Abstract
The development of novel approaches to control immune responses to self- and allogenic tissues/organs represents an ambitious goal for the management of autoimmune diseases and in transplantation. Regulatory T cells (Tregs) are recognized as key players in the maintenance of peripheral tolerance in physiological and pathological conditions, and Treg-based cell therapies to restore tolerance in T cell-mediated disorders have been designed. However, several hurdles, including insufficient number of Tregs, their stability, and their antigen specificity, have challenged Tregs clinical applicability. In the past decade, the ability to engineer T cells has proven a powerful tool to redirect specificity and function of different cell types for specific therapeutic purposes. By using lentivirus-mediated gene transfer of the thymic-derived Treg transcription factor forkhead-box-P3 (FOXP3) in conventional CD4+ T cells, we converted effector T cells into Treg-like cells, endowed with potent in vitro and in vivo suppressive activity. The resulting CD4FOXP3 T-cell population displays stable phenotype and suppressive function. We showed that this strategy restores Treg function in T lymphocytes from patients carrying mutations in FOXP3 [immune-dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX)], in whom CD4FOXP3 T cell could be used as therapeutics to control autoimmunity. Here, we will discuss the potential advantages of using CD4FOXP3 T cells for in vivo application in inflammatory diseases, where tissue inflammation may undermine the function of natural Tregs. These findings pave the way for the use of engineered Tregs not only in IPEX syndrome but also in autoimmune disorders of different origin and in the context of stem cell and organ transplantation.
Collapse
Affiliation(s)
- Laura Passerini
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosa Bacchetta
- Department of Stem Cell Transplantation and Regenerative Medicine, Division of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
| |
Collapse
|
140
|
Hull CM, Peakman M, Tree TIM. Regulatory T cell dysfunction in type 1 diabetes: what's broken and how can we fix it? Diabetologia 2017; 60:1839-1850. [PMID: 28770318 PMCID: PMC6448885 DOI: 10.1007/s00125-017-4377-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/17/2017] [Indexed: 01/07/2023]
Abstract
Type 1 diabetes is an autoimmune disease characterised by the destruction of insulin producing beta cells in the pancreas. Whilst it remains unclear what the original triggering factors for this destruction are, observations from the natural history of human type 1 diabetes, including incidence rates in twins, suggest that the disease results from a combination of genetic and environmental factors. Whilst many different immune cells have been implicated, including members of the innate and adaptive immune systems, a view has emerged over the past 10 years that beta cell damage is mediated by the combined actions of CD4+ and CD8+ T cells with specificity for islet autoantigens. In health, these potentially pathogenic T cells are held in check by multiple regulatory mechanisms, known collectively as 'immunological tolerance'. This raises the question as to whether type 1 diabetes develops, at least in part, as a result of a defect in one or more of these control mechanisms. Immunological tolerance includes both central mechanisms (purging of the T cell repertoire of high-affinity autoreactive T cells in the thymus) and peripheral mechanisms, a major component of which is the action of a specialised subpopulation of T cells, known as regulatory T cells (Tregs). In this review, we highlight the evidence suggesting that a reduction in the functional capacity of different Treg populations contributes to disease development in type 1 diabetes. We also address current controversies regarding the putative causes of this defect and discuss strategies to correct it as a means to reduce or prevent islet destruction in a clinical setting.
Collapse
Affiliation(s)
- Caroline M Hull
- Programme of Infection and Immunity, Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, Borough Wing, Guy's Hospital, London, SE1 9RT, UK.
| | - Mark Peakman
- Programme of Infection and Immunity, Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Timothy I M Tree
- Programme of Infection and Immunity, Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, Borough Wing, Guy's Hospital, London, SE1 9RT, UK.
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK.
| |
Collapse
|
141
|
Abstract
Precision medicine is an emerging integrative approach for disease prevention, early detection, and treatment which takes into account individual variability in genetic and other molecular measurements, medical history, environmental exposures, and lifestyle. The development and availability of genomic and other molecular profiling technologies provide an unprecedented opportunity to apply precision medicine strategies in transplantation research. Developing integrative computational methods to analyze these diverse types of data provides new opportunities to impact diagnostics and therapeutics. In this article, we discuss ways we can leverage molecular data sets to develop new hypotheses for disease mechanisms, identify new disease biomarkers, and reposition drugs for diseases with unmet needs. We specifically discuss computational methods that can be applied to achieve these goals in the context of organ transplant.
Collapse
|
142
|
Delalat B, Harding F, Gundsambuu B, De-Juan-Pardo EM, Wunner FM, Wille ML, Jasieniak M, Malatesta KA, Griesser HJ, Simula A, Hutmacher DW, Voelcker NH, Barry SC. 3D printed lattices as an activation and expansion platform for T cell therapy. Biomaterials 2017. [DOI: 10.1016/j.biomaterials.2017.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
143
|
Dawson NAJ, Levings MK. Antigen-specific regulatory T cells: are police CARs the answer? Transl Res 2017; 187:53-58. [PMID: 28688236 DOI: 10.1016/j.trsl.2017.06.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/05/2017] [Accepted: 06/13/2017] [Indexed: 01/26/2023]
Abstract
Cellular therapy with T-regulatory cells (Tregs) is a promising strategy to control immune responses and restore immune tolerance in a variety of immune-mediated diseases, such as transplant rejection and autoimmunity. Multiple clinical trials are currently testing this approach, typically by infusing a single dose of polyclonal Tregs that have been expanded in vitro. However, evidence from animal models of Treg therapy has clearly shown that antigen-specific Tregs are vastly superior to polyclonal cells, meaning that fewer cells are needed for the desired therapeutic effect. Traditional methods to obtain antigen-specific Tregs include antigen-stimulated expansion or T-cell receptor (TCR) overexpression. However, these methods are limited by low cell numbers, complex manufacturing procedures, and knowledge of patient-specific TCRs which recognize disease-relevant MHC-peptide complexes. Recently, several groups have explored the potential to use chimeric antigen receptors (CARs) to generate antigen-specific Tregs. Here, we discuss the progress in this field and highlight the major outstanding questions that remain to be addressed as this approach moves toward clinical applications.
Collapse
Affiliation(s)
- Nicholas A J Dawson
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia & British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
144
|
Alharshawi K, Marinelarena A, Kumar P, El-Sayed O, Bhattacharya P, Sun Z, Epstein AL, Maker AV, Prabhakar BS. PKC-ѳ is dispensable for OX40L-induced TCR-independent Treg proliferation but contributes by enabling IL-2 production from effector T-cells. Sci Rep 2017; 7:6594. [PMID: 28747670 PMCID: PMC5529425 DOI: 10.1038/s41598-017-05254-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/25/2017] [Indexed: 01/07/2023] Open
Abstract
We have previously shown that OX40L/OX40 interaction is critical for TCR-independent selective proliferation of Foxp3+ Tregs, but not Foxp3- effector T-cells (Teff), when CD4+ T-cells are co-cultured with GM-CSF derived bone marrow dendritic cells (G-BMDCs). Events downstream of OX40L/OX40 interaction in Tregs responsible for this novel mechanism are not understood. Earlier, OX40L/OX40 interaction has been shown to stimulate CD4+ T-cells through the formation of a signalosome involving TRAF2/PKC-Ѳ leading to NF-kB activation. In this study, using CD4+ T-cells from WT and OX40-/- mice we first established that OX40 mediated activation of NF-kB was critical for this Treg proliferation. Although CD4+ T-cells from PKC-Ѳ-/- mice were also defective in G-BMDC induced Treg proliferation ex vivo, this defect could be readily corrected by adding exogenous IL-2 to the co-cultures. Furthermore, by treating WT, OX40-/-, and PKC-Ѳ-/- mice with soluble OX40L we established that OX40L/OX40 interaction was required and sufficient to induce Treg proliferation in vivo independent of PKC-Ѳ status. Although PKC-Ѳ is dispensable for TCR-independent Treg proliferation per se, it is essential for optimum IL-2 production by Teff cells. Finally, our findings suggest that OX40L binding to OX40 likely results in recruitment of TRAF1 for downstream signalling.
Collapse
Affiliation(s)
- Khaled Alharshawi
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Alejandra Marinelarena
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Osama El-Sayed
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Palash Bhattacharya
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zuoming Sun
- Department of Immunology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Alan L Epstein
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Ajay V Maker
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA.,Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
145
|
Abstract
Modern immunosuppression regimens effectively control acute rejection and decrease graft loss in the first year after transplantation; however, these regimens do not have a durable effect on long-term graft survival owing to a combination of drug toxicities and the emergence of chronic alloimmune responses. Eliminating drugs and their toxicities while maintaining graft acceptance has been the primary aim of cellular therapies. Tregs suppress both autoimmune and alloimmune responses and are particularly effective in protecting allografts in experimental transplant models. Further, Treg-based therapies are selective, do not require harsh conditioning, and do not have a risk of graft-versus-host disease. Trial designs should consider the distinct immunological features of each transplanted organ, Treg preparations, dose, and frequency, and the ability to detect and quantify Treg effects in a given transplant environment. In this Review, we detail the ongoing clinical trials of Treg therapy in liver and kidney transplantation. Integration of Treg biology gleaned from preclinical models and experiences in human organ transplantation should allow for optimization of trial design that will determine the potential efficacy of a given therapy and provide guidelines for further therapeutic development.
Collapse
Affiliation(s)
- Qizhi Tang
- Department of Surgery.,Diabetes Center, and
| | - Flavio Vincenti
- Department of Surgery.,Department of Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
146
|
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: 56] [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
|
147
|
Donor-Reactive Regulatory T Cell Frequency Increases During Acute Cellular Rejection of Lung Allografts. Transplantation 2017; 100:2090-8. [PMID: 27077597 DOI: 10.1097/tp.0000000000001191] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Acute cellular rejection is a major cause of morbidity after lung transplantation. Because regulatory T (Treg) cells limit rejection of solid organs, we hypothesized that donor-reactive Treg increase after transplantation with development of partial tolerance and decrease relative to conventional CD4 (Tconv) and CD8 T cells during acute cellular rejection. METHODS To test these hypotheses, we prospectively collected 177 peripheral blood mononuclear cell specimens from 39 lung transplant recipients at the time of transplantation and during bronchoscopic assessments for acute cellular rejection. We quantified the proportion of Treg, CD4 Tconv, and CD8 T cells proliferating in response to donor-derived, stimulated B cells. We used generalized estimating equation-adjusted regression to compare donor-reactive T cell frequencies with acute cellular rejection pathology. RESULTS An average of 16.5 ± 9.0% of pretransplantation peripheral blood mononuclear cell Treg cell were donor-reactive, compared with 3.8% ± 2.9% of CD4 Tconv and 3.4 ± 2.6% of CD8 T cells. These values were largely unchanged after transplantation. Donor-reactive CD4 Tconv and CD8 T cell frequencies both increased 1.5-fold (95% confidence interval [95% CI], 1.3-1.6; P < 0.001 and 95% CI, 1.2-1.6; P = 0.007, respectively) during grade A2 rejection compared with no rejection. Surprisingly, donor-reactive Treg frequencies increased by 1.7-fold (95% CI, 1.4-1.8; P < 0.001). CONCLUSIONS Contrary to prediction, overall proportions of donor-reactive Treg cells are similar before and after transplantation and increase during grade A2 rejection. This suggests how A2 rejection can be self-limiting. The observed increases over high baseline proportions in donor-reactive Treg were insufficient to prevent acute lung allograft rejection.
Collapse
|
148
|
Wong TC, Lo CM, Fung JY. Emerging drugs for prevention of T-cell mediated rejection in liver and kidney transplantation. Expert Opin Emerg Drugs 2017; 22:123-136. [PMID: 28503959 DOI: 10.1080/14728214.2017.1330884] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Acute and chronic graft rejection continues to be an important problem after solid organ transplantation. With the introduction of potent immunosuppressive agents such as calcineurin inhibitors, the risk of rejection has been significantly reduced. However, the adverse effects of life-long immunosuppression remain a concern, and there exist a fine balance between over-immunosuppression and risk of rejection. Areas covered: In this review, the current standard of care in immunosuppressive therapy, including the use of steroids, calcineurin inhibitors, mycophenolate prodrugs and mammalian target of rapamycin inhibitors, will be discussed. Newer immunosuppressive agents showing promising early data after liver and kidney transplantation will also be explored. Expert Opinion: Currently, calcineurin inhibitors continue to be a vital component of immunosuppressive therapy after solid organ transplantation. Although minimization and avoidance strategies have been developed, the ultimate goal of inducing tolerance remains elusive. Newer emerging agents should have potent and specific immunosuppressive activity, with minimal associated side effects. An individualized approach should be adopted to tailor immunosuppression according to the different needs of recipients.
Collapse
Affiliation(s)
- Tiffany Cl Wong
- a Department of Surgery, Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong , Hong Kong S.A.R
| | - Chung-Mau Lo
- a Department of Surgery, Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong , Hong Kong S.A.R
| | - James Yy Fung
- a Department of Surgery, Department of Medicine , Queen Mary Hospital, The University of Hong Kong , Hong Kong , Hong Kong S.A.R
| |
Collapse
|
149
|
Zwang NA, Leventhal JR. Cell Therapy in Kidney Transplantation: Focus on Regulatory T Cells. J Am Soc Nephrol 2017; 28:1960-1972. [PMID: 28465379 DOI: 10.1681/asn.2016111206] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Renal transplantation is the renal replacement modality of choice for suitable candidates with advanced CKD or ESRD. Prevention of rejection, however, requires treatment with nonspecific pharmacologic immunosuppressants that carry both systemic and nephrologic toxicities. Use of a patient's own suppressive regulatory T cells (Tregs) is an attractive biologic approach to reduce this burden. Here, we review the immunologic underpinnings of Treg therapy and technical challenges to developing successful cell therapy. These issues include the selection of appropriate Treg subsets, ex vivo Treg expansion approaches, how many Tregs to administer and when, and how to care for patients after Treg administration.
Collapse
Affiliation(s)
| | - Joseph R Leventhal
- Comprehensive Transplant Center, Northwestern Memorial Hospital, Chicago, Illinois
| |
Collapse
|
150
|
Abstract
Type 1 diabetes is an autoimmune disorder in which the immune system attacks and destroys insulin-producing islet cells of the pancreas. Although islet transplantation has proved to be successful for some patients with type 1 diabetes, its widespread use is limited by islet donor shortage and the requirement for lifelong immunosuppression. An encapsulation strategy that can prevent the rejection of xenogeneic islets or of stem cell-derived allogeneic islets can potentially eliminate both of these barriers. Although encapsulation technology has met several challenges, the convergence of expertise in materials, nanotechnology, stem cell biology and immunology is allowing us to get closer to the goal of encapsulated islet cell therapy for humans.
Collapse
Affiliation(s)
- Tejal Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, Byers Hall Rm 203C, MC 2520, 1700 4th Street, San Francisco, California 94158-2330, USA
| | - Lonnie D Shea
- University of Michigan, Department of Biomedical Engineering, 1119 Carl A. Gerstacker Building, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109-2099, USA
| |
Collapse
|