151
|
Hull CM, Nickolay LE, Estorninho M, Richardson MW, Riley JL, Peakman M, Maher J, Tree TI. Generation of human islet-specific regulatory T cells by TCR gene transfer. J Autoimmun 2017; 79:63-73. [DOI: 10.1016/j.jaut.2017.01.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 01/12/2023]
|
152
|
Noyan F, Zimmermann K, Hardtke-Wolenski M, Knoefel A, Schulde E, Geffers R, Hust M, Huehn J, Galla M, Morgan M, Jokuszies A, Manns MP, Jaeckel E. Prevention of Allograft Rejection by Use of Regulatory T Cells With an MHC-Specific Chimeric Antigen Receptor. Am J Transplant 2017; 17:917-930. [PMID: 27997080 DOI: 10.1111/ajt.14175] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 01/25/2023]
Abstract
CD4+ CD25high FOXP3+ regulatory T cells (Tregs) are involved in graft-specific tolerance after solid organ transplantation. However, adoptive transfer of polyspecific Tregs alone is insufficient to prevent graft rejection even in rodent models, indicating that graft-specific Tregs are required. We developed a highly specific chimeric antigen receptor that recognizes the HLA molecule A*02 (referred to as A2-CAR). Transduction into natural regulatory T cells (nTregs) changes the specificity of the nTregs without alteration of their regulatory phenotype and epigenetic stability. Activation of nTregs via the A2-CAR induced proliferation and enhanced the suppressor function of modified nTregs. Compared with nTregs, A2-CAR Tregs exhibited superior control of strong allospecific immune responses in vitro and in humanized mouse models. A2-CAR Tregs completely prevented rejection of allogeneic target cells and tissues in immune reconstituted humanized mice in the absence of any immunosuppression. Therefore, these modified cells have great potential for incorporation into clinical trials of Treg-supported weaning after allogeneic transplantation.
Collapse
Affiliation(s)
- F Noyan
- Department of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany.,Integrated Research and Treatment Center, Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| | - K Zimmermann
- Department of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - M Hardtke-Wolenski
- Department of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - A Knoefel
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - E Schulde
- Department of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - R Geffers
- RG of Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - M Hust
- Department of Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - J Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - M Galla
- Institute of Experimental Haematology, Hannover Medical School, Hannover, Germany
| | - M Morgan
- Institute of Experimental Haematology, Hannover Medical School, Hannover, Germany
| | - A Jokuszies
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - M P Manns
- Department of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - E Jaeckel
- Department of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany.,Integrated Research and Treatment Center, Transplantation (IFB-Tx), Hannover Medical School, Hannover, Germany
| |
Collapse
|
153
|
Le Guen V, Judor JP, Boeffard F, Gauttier V, Ferry N, Soulillou JP, Brouard S, Conchon S. Alloantigen gene transfer to hepatocytes promotes tolerance to pancreatic islet graft by inducing CD8 + regulatory T cells. J Hepatol 2017; 66:765-777. [PMID: 27914923 DOI: 10.1016/j.jhep.2016.11.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 11/10/2016] [Accepted: 11/13/2016] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Induction of donor-specific immune tolerance is a good alternative to chronic life-long immunosuppression for transplant patients. Donor major histocompatibility complex (MHC) molecules represent the main targets of the allogeneic immune response of transplant recipients. Liver targeted gene transfer with viral vectors induces tolerance toward the encoded antigen. The aim of this work was to determine whether alloantigen gene transfer to hepatocytes induces tolerance and promotes graft acceptance. METHODS C57BL/6 (H-2b) mice were treated with adeno-associated viral (AAV) vector targeting the expression of the MHC class I molecule H-2Kd to hepatocytes, before transplantation with fully allogeneic pancreatic islet from BALB/c mice (H-2d). RESULTS AAV H-2Kd treated mice were tolerant to the alloantigen, as demonstrated by its long-term expression by the hepatocytes, even after a highly immunogenic challenge with an adenoviral vector. After chemical induction of diabetes, the AAV treated mice had significantly delayed rejection of fully allogeneic pancreatic islet grafts, with more than 40% of recipients tolerant (>100days). AAV-mediated expression of H-2Kd in the liver induced the local expansion of CD8+ T lymphocytes with allo-specific suppressive properties. The adoptive transfer of these liver-generated CD8+ Tregs into naive diabetic mice promoted the long-term survival of allogeneic pancreatic islet grafts. CONCLUSION AAV-mediated long-term expression of a single MHC class I molecule in the liver induces the generation of a subset of allo-specific CD8+ Treg cells, which promote tolerance toward fully allogeneic graft. Liver gene transfer represents a promising strategy for in vivo induction of donor-specific tolerance. LAY SUMMARY The liver has a special immune system, biased toward tolerance. In this study, we investigated the possibility of harnessing this property of the liver to induce tolerance to an allogeneic transplantation. We demonstrate for the first time that the in vivo gene transfer of an allogeneic antigen with an adeno-associated viral vector to mouse hepatocytes induces the expansion of a population of CD8+ regulatory T lymphocytes. These Tregs are then instrumental in preventing the rejection of allogeneic pancreatic islets transplanted in these animals. Allogeneic transplantation is the main treatment for the end-stage diseases of a number of organs. Life-long immunosuppressive treatments are still required to limit graft rejection, and these treatments exhibit serious side effects. Our present findings open a new avenue for promoting allo-specific tolerance via in vivo induction of CD8+ Treg expansion.
Collapse
Affiliation(s)
- Valentin Le Guen
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Jean-Paul Judor
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Françoise Boeffard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Vanessa Gauttier
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Nicolas Ferry
- Département de Thérapie Cellulaire, CHU Saint Louis, Paris, France
| | - Jean-Paul Soulillou
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Sophie Conchon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France; Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.
| |
Collapse
|
154
|
Boardman DA, Philippeos C, Fruhwirth GO, Ibrahim MAA, Hannen RF, Cooper D, Marelli-Berg FM, Watt FM, Lechler RI, Maher J, Smyth LA, Lombardi G. Expression of a Chimeric Antigen Receptor Specific for Donor HLA Class I Enhances the Potency of Human Regulatory T Cells in Preventing Human Skin Transplant Rejection. Am J Transplant 2017; 17:931-943. [PMID: 28027623 DOI: 10.1111/ajt.14185] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/30/2016] [Accepted: 12/17/2016] [Indexed: 01/25/2023]
Abstract
Regulatory T cell (Treg) therapy using recipient-derived Tregs expanded ex vivo is currently being investigated clinically by us and others as a means of reducing allograft rejection following organ transplantation. Data from animal models has demonstrated that adoptive transfer of allospecific Tregs offers greater protection from graft rejection compared to polyclonal Tregs. Chimeric antigen receptors (CAR) are clinically translatable synthetic fusion proteins that can redirect the specificity of T cells toward designated antigens. We used CAR technology to redirect human polyclonal Tregs toward donor-MHC class I molecules, which are ubiquitously expressed in allografts. Two novel HLA-A2-specific CARs were engineered: one comprising a CD28-CD3ζ signaling domain (CAR) and one lacking an intracellular signaling domain (ΔCAR). CAR Tregs were specifically activated and significantly more suppressive than polyclonal or ΔCAR Tregs in the presence of HLA-A2, without eliciting cytotoxic activity. Furthermore, CAR and ΔCAR Tregs preferentially transmigrated across HLA-A2-expressing endothelial cell monolayers. In a human skin xenograft transplant model, adoptive transfer of CAR Tregs alleviated the alloimmune-mediated skin injury caused by transferring allogeneic peripheral blood mononuclear cells more effectively than polyclonal Tregs. Our results demonstrated that the use of CAR technology is a clinically applicable refinement of Treg therapy for organ transplantation.
Collapse
Affiliation(s)
- D A Boardman
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK
| | - C Philippeos
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - G O Fruhwirth
- Department of Imaging Chemistry and Biology, Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas' Hospital, London, UK
| | - M A A Ibrahim
- Department of Clinical Immunology and Allergy, King's College London, King's College Hospital, London, UK.,Division of Asthma, Allergy & Lung Biology, King's College London, Guy's Hospital, London, UK
| | - R F Hannen
- Centre for Cell Biology & Cutaneous Research, Bart's and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - D Cooper
- William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, London, UK
| | - F M Marelli-Berg
- William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, London, UK
| | - F M Watt
- NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK.,Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - R I Lechler
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK
| | - J Maher
- NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK.,Department of Clinical Immunology and Allergy, King's College London, King's College Hospital, London, UK.,CAR Mechanics Group, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| | - L A Smyth
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,School of Health, Sport and Bioscience, Stratford Campus, University of East London, London, UK
| | - G Lombardi
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK
| |
Collapse
|
155
|
Ferreira LMR, Tang Q. Generating Antigen-Specific Regulatory T Cells in the Fast Lane. Am J Transplant 2017; 17:851-853. [PMID: 28102024 DOI: 10.1111/ajt.14202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 12/23/2016] [Accepted: 01/08/2017] [Indexed: 01/25/2023]
Affiliation(s)
- L M R Ferreira
- Department of Surgery, University of California, San Francisco, San Francisco, CA.,UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Q Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA.,UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
156
|
Romano M, Tung SL, Smyth LA, Lombardi G. Treg therapy in transplantation: a general overview. Transpl Int 2017; 30:745-753. [PMID: 28012226 DOI: 10.1111/tri.12909] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/26/2016] [Accepted: 12/19/2016] [Indexed: 12/23/2022]
Abstract
Solid organ transplantation remains the treatment of choice for end-stage organ failure. Whilst the short-term outcomes post-transplant have improved in the last decades, chronic rejection and immunosuppressant side effects remain an ongoing concern. Hematopoietic stem cell transplantation is a well-established procedure for the treatment of patients with haematological disorders. However, donor T cells are continually primed and activated to react against the host causing graft-versus-host disease (GvHD) that leads to tissue damages and death. Regulatory T cells (Tregs) play an essential role in maintaining tolerance to self-antigens, preventing excessive immune responses and abrogating autoimmunity. Due to their suppressive properties, Tregs have been extensively studied for their use as a cellular therapy aiming to treat GvHD and limit immune responses responsible for graft rejection. Several clinical trials have been conducted or are currently ongoing to investigate safety and feasibility of Treg-based therapy. This review summarizes the general understanding of Treg biology and presents the methods used to isolate and expand Tregs. Furthermore, we describe data from the first clinical trials using Tregs, explaining the limitations and future application of these cells.
Collapse
Affiliation(s)
- Marco Romano
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Sim Lai Tung
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Lesley Ann Smyth
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,School of Health Sport and Bioscience, University of East London, London, UK
| | - Giovanna Lombardi
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
157
|
Esensten JH, Bluestone JA, Lim WA. Engineering Therapeutic T Cells: From Synthetic Biology to Clinical Trials. ANNUAL REVIEW OF PATHOLOGY 2017; 12:305-330. [PMID: 27959633 PMCID: PMC5557092 DOI: 10.1146/annurev-pathol-052016-100304] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Engineered T cells are currently in clinical trials to treat patients with cancer, solid organ transplants, and autoimmune diseases. However, the field is still in its infancy. The design, and manufacturing, of T cell therapies is not standardized and is performed mostly in academic settings by competing groups. Reliable methods to define dose and pharmacokinetics of T cell therapies need to be developed. As of mid-2016, there are no US Food and Drug Administration (FDA)-approved T cell therapeutics on the market, and FDA regulations are only slowly adapting to the new technologies. Further development of engineered T cell therapies requires advances in immunology, synthetic biology, manufacturing processes, and government regulation. In this review, we outline some of these challenges and discuss the contributions that pathologists can make to this emerging field.
Collapse
Affiliation(s)
- Jonathan H Esensten
- Department of Laboratory Medicine, University of California, San Francisco, California 94143;
| | - Jeffrey A Bluestone
- Diabetes Center and Department of Medicine, University of California, San Francisco, California 94143;
| | - Wendell A Lim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco 94158-2517;
| |
Collapse
|
158
|
Ghali JR, Wang YM, Holdsworth SR, Kitching AR. Regulatory T cells in immune-mediated renal disease. Nephrology (Carlton) 2016. [PMID: 26206106 DOI: 10.1111/nep.12574] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Regulatory T cells (Tregs) are CD4+ T cells that can suppress immune responses by effector T cells, B cells and innate immune cells. This review discusses the role that Tregs play in murine models of immune-mediated renal diseases and acute kidney injury and in human autoimmune kidney disease (such as systemic lupus erythematosus, anti-glomerular basement membrane disease, anti-neutrophil cytoplasmic antibody-associated vasculitis). Current research suggests that Tregs may be reduced in number and/or have impaired regulatory function in these diseases. Tregs possess several mechanisms by which they can limit renal and systemic inflammatory immune responses. Potential therapeutic applications involving Tregs include in vivo induction of Tregs or inducing Tregs from naïve CD4+ T cells or expanding natural Tregs ex vivo, to use as a cellular therapy. At present, the optimal method of generating a phenotypically stable pool of Tregs with long-lasting suppressive effects is not established, but human studies in renal transplantation are underway exploring the therapeutic potential of Tregs as a cellular therapy, and if successful may have a role as a novel therapy in immune-mediated renal diseases.
Collapse
Affiliation(s)
- Joanna R Ghali
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Victoria.,Department of Nephrology, Monash Medical Centre, Melbourne, Victoria
| | - Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Victoria.,Department of Nephrology, Monash Medical Centre, Melbourne, Victoria
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Victoria.,Department of Nephrology, Monash Medical Centre, Melbourne, Victoria.,Department of Paediatric Nephrology, Monash Medical Centre, Melbourne, Victoria
| |
Collapse
|
159
|
Safinia N, Vaikunthanathan T, Fraser H, Thirkell S, Lowe K, Blackmore L, Whitehouse G, Martinez-Llordella M, Jassem W, Sanchez-Fueyo A, Lechler RI, Lombardi G. Successful expansion of functional and stable regulatory T cells for immunotherapy in liver transplantation. Oncotarget 2016; 7:7563-77. [PMID: 26788992 PMCID: PMC4884938 DOI: 10.18632/oncotarget.6927] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 01/01/2016] [Indexed: 12/14/2022] Open
Abstract
Strategies to prevent organ transplant rejection whilst minimizing long-term immunosuppression are currently under intense investigation with regulatory T cells (Tregs) nearing clinical application. The clinical trial, ThRIL, recently commenced at King's College London, proposes to use Treg cell therapy to induce tolerance in liver transplant recipients, the success of which has the potential to revolutionize the management of these patients and enable a future of drug-free transplants. This is the first report of the manufacture of clinical grade Tregs from prospective liver transplant recipients via a CliniMACS-based GMP isolation technique and expanded using anti-CD3/CD28 beads, IL-2 and rapamycin. We report the enrichment of a pure, stable population of Tregs (>95% CD4(+)CD25(+)FOXP3(+)), reaching adequate numbers for their clinical application. Our protocol proved successful in, influencing the expansion of superior functional Tregs, as compared to freshly isolated cells, whilst also preventing their conversion to Th17 cells under pro-inflammatory conditions. We conclude with the manufacture of the final Treg product in the clinical research facility (CRF), a prerequisite for the clinical application of these cells. The data presented in this manuscript together with the much-anticipated clinical results from ThRIL, will undoubtedly inform the improved management of the liver transplant recipient.
Collapse
Affiliation(s)
- Niloufar Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| | - Trishan Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| | - Henrieta Fraser
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| | - Sarah Thirkell
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| | - Katie Lowe
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| | - Laura Blackmore
- Institute of Liver Studies, King's College Hospital, London, UK
| | | | | | - Wayel Jassem
- Institute of Liver Studies, King's College Hospital, London, UK
| | | | - Robert I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
160
|
Litjens NHR, Boer K, Zuijderwijk JM, Klepper M, Peeters AMA, Verschoor W, Kraaijeveld R, Betjes MGH. Natural regulatory T cells from patients with end-stage renal disease can be used for large-scale generation of highly suppressive alloantigen-specific Tregs. Kidney Int 2016; 91:1203-1213. [PMID: 27988212 DOI: 10.1016/j.kint.2016.09.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/22/2016] [Accepted: 09/29/2016] [Indexed: 10/20/2022]
Abstract
Natural occurring regulatory T cells (nTregs) have the potential to offer a targeted approach of immunosuppression and are the cell type of interest for inducing tolerance in kidney transplantation. End-stage renal disease (ESRD) profoundly affects the composition and function of circulating T cells but little is known with respect to how nTreg potential is affected. To address this, nTregs of patients with ESRD (on dialysis or not) and healthy individuals were isolated, expanded using allogeneic mature monocyte-derived dendritic cells followed by anti-CD3/anti-CD28-coated beads and the different nTregs were extensively characterized by the demethylation status of the Treg-specific demethylated region within FOXP3 and expression of typical nTreg markers. Additionally, the suppressive capacity as well as cytokine producing cells were analyzed for allogeneic mature monocyte-derived dendritic cell-expanded nTregs. Compared to age- and gender-matched healthy individuals, similar frequencies of nTregs were present within the circulation of patients with ESRD either on dialysis or not. The isolated nTregs could be equally well or even better expanded using allogeneic mature monocyte-derived dendritic cells and extensive characterization did not reveal significant differences. The demethylation status of the Treg-specific demethylated region was maintained or even further promoted as was the expression of markers characteristic for nTregs. Moreover, suppressive capacity and the cytokine profile of allogeneic mature monocyte-derived dendritic cell-expanded nTregs was similar to that of healthy individuals. Thus, circulating nTregs of patients with ESRD can effectively be expanded to stable allo antigen-specific nTregs with potential clinical applicability.
Collapse
Affiliation(s)
- Nicolle H R Litjens
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Karin Boer
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joke M Zuijderwijk
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Mariska Klepper
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Annemiek M A Peeters
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wenda Verschoor
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rens Kraaijeveld
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
161
|
Ezzelarab MB, Thomson AW. Adoptive Cell Therapy with Tregs to Improve Transplant Outcomes: The Promise and the Stumbling Blocks. CURRENT TRANSPLANTATION REPORTS 2016; 3:265-274. [PMID: 28529840 PMCID: PMC5435383 DOI: 10.1007/s40472-016-0114-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The contribution of regulatory T cells (Treg) to the induction and maintenance of tolerance is well-recognized in rodents and may contribute to long-term human organ allograft survival. The therapeutic efficacy of adoptively-transferred Treg in promoting tolerance to organ allografts is well-recognized in mouse models. Early phase 1/2 clinical studies of Treg therapy have been conducted in patients with type-1 (autoimmune) diabetes and refractory Crohn's disease, and for inhibition of graft-versus-host disease following bone marrow transplantation with proven safety. The feasibility of adoptive Treg therapy in the clinic is subject to various parameters, including optimal cell source, isolation procedure, expansion, target dose, time of infusion, as well as generation of a GMP-cell product. Several phase 1/2 Treg dose-escalation studies are underway in organ transplantation. Recent evidence suggests that additional factors are critical to ensure Treg safety and efficacy in allograft recipients, including Treg characterization, stability, longevity, trafficking, concomitant immunosuppression, and donor antigen specificity. Accordingly, Treg therapy in the context of organ transplantation may prove more challenging in comparison to other prospective clinical settings of Treg immunotherapy, such as type-1 diabetes.
Collapse
Affiliation(s)
- Mohamed B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Angus W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
162
|
|
163
|
Theil A, Wilhelm C, Kuhn M, Petzold A, Tuve S, Oelschlägel U, Dahl A, Bornhäuser M, Bonifacio E, Eugster A. T cell receptor repertoires after adoptive transfer of expanded allogeneic regulatory T cells. Clin Exp Immunol 2016; 187:316-324. [PMID: 27774628 DOI: 10.1111/cei.12887] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2016] [Indexed: 12/16/2022] Open
Abstract
Regulatory T cell (Treg ) therapy has been exploited in autoimmune disease, solid organ transplantation and in efforts to prevent or treat graft-versus-host disease (GVHD). However, our knowledge on the in-vivo persistence of transfused Treg is limited. Whether Treg transfusion leads to notable changes in the overall Treg repertoire or whether longevity of Treg in the periphery is restricted to certain clones is unknown. Here we use T cell receptor alpha chain sequencing (TCR-α-NGS) to monitor changes in the repertoire of Treg upon polyclonal expansion and after subsequent adoptive transfer. We applied TCR-α-NGS to samples from two patients with chronic GVHD who received comparable doses of stem cell donor derived expanded Treg . We found that in-vitro polyclonal expansion led to notable repertoire changes in vitro and that Treg cell therapy altered the peripheral Treg repertoire considerably towards that of the infused cell product, to different degrees, in each patient. Clonal changes in the peripheral blood were transient and correlated well with the clinical parameters. We suggest that T cell clonotype analyses using TCR sequencing should be considered as a means to monitor longevity and fate of adoptively transferred T cells.
Collapse
Affiliation(s)
- A Theil
- DFG-Center for Regenerative Therapies Dresden, Dresden, Germany.,Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - C Wilhelm
- DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| | - M Kuhn
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Dresden, Germany
| | - A Petzold
- Deep Sequencing Group, Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - S Tuve
- Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - U Oelschlägel
- Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - A Dahl
- Deep Sequencing Group, Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - M Bornhäuser
- Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - E Bonifacio
- DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| | - A Eugster
- DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
164
|
Dargaud Y, Pavlova A, Lacroix-Desmazes S, Fischer K, Soucie M, Claeyssens S, Scott DW, d'Oiron R, Lavigne-Lissalde G, Kenet G, Escuriola Ettingshausen C, Borel-Derlon A, Lambert T, Pasta G, Négrier C. Achievements, challenges and unmet needs for haemophilia patients with inhibitors: Report from a symposium in Paris, France on 20 November 2014. Haemophilia 2016; 22 Suppl 1:1-24. [PMID: 26728503 DOI: 10.1111/hae.12860] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2015] [Indexed: 12/28/2022]
Abstract
Over the past 20 years, there have been many advances in haemophilia treatment that have allowed patients to take greater control of their disease. However, the development of factor VIII (FVIII) inhibitors is the greatest complication of the disease and a challenge in the treatment of haemophilia making management of bleeding episodes difficult and surgical procedures very challenging. A meeting to discuss the unmet needs of haemophilia patients with inhibitors was held in Paris on 20 November 2014. Topics discussed were genetic and non-genetic risk factors for the development of inhibitors, immunological aspects of inhibitor development, FVIII products and inhibitor development, generation and functional properties of engineered antigen-specific T regulatory cells, suppression of immune responses to FVIII, prophylaxis in haemophilia patients with inhibitors, epitope mapping of FVIII inhibitors, current controversies in immune tolerance induction therapy, surgery in haemophilia patients with inhibitors and future perspectives for the treatment of haemophilia patients with inhibitors. A summary of the key points discussed is presented in this paper.
Collapse
Affiliation(s)
- Y Dargaud
- Unite d'Hemostase Clinique, Hopital Cardiologique Louis Pradel, Universite Lyon 1, Lyon, France
| | - A Pavlova
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic, Bonn, Germany
| | - S Lacroix-Desmazes
- INSERM UMRS 1138, Immunopathologie et immuno-intervention thérapeutique, Centre de Recherche des Cordeliers, Paris, France
| | - K Fischer
- Van Creveldkliniek (HP C01.425), University Medical Center Utrecht, Utrecht, The Netherlands
| | - M Soucie
- Division of Blood Disorders, National Center for Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - S Claeyssens
- Chu Purpan Pav. Centre Hospitalier Lefebvre, Centre Rgal de l'Hemophilie, Toulouse, France
| | - D W Scott
- Department of Medicine, Uniformed Services, University for the Health Sciences, Bethesda, MD, USA
| | - R d'Oiron
- Centre de Traitement de l'Hémophilie et des Maladies Hémorragiques Constitutionnelles, Hôpitaux Universitaires Paris Sud - Site Bicêtre, Le Kremlin-Bicêtre, France
| | - G Lavigne-Lissalde
- Laboratoire d'Hématologie et Consultations d'Hématologie Biologique Centre Hospitalier Universitaire de Nîmes, Place du Pr R. Debré Nîmes, France
| | - G Kenet
- National Hemophilia Institute, Sheba Medical Center, Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | | | - A Borel-Derlon
- Haemophilia and von Willebrand Disease Centre, University Hospital of Caen, Caen
| | - T Lambert
- Hemophilia Care Center, Bicêtre AP-HP Hospital and Faculté de Médecine Paris XI, Paris, France
| | - G Pasta
- UOSD di Ortopedia e Traumatologia, Centro Emofilia 'Angelo Bianchi Bonomi', Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - C Négrier
- Haematology Department, Director Hemophilia Comprehensive Care Center, Hopital Louis Pradel, Université Lyon 1, Bron Cedex, France
| |
Collapse
|
165
|
Abstract
Direct allorecognition is the process by which donor-derived major histocompatibility complex (MHC)-peptide complexes, typically presented by donor-derived ‘passenger’ dendritic cells, are recognised directly by recipient T cells. In this review, we discuss the two principle theories which have been proposed to explain why individuals possess a high-precursor frequency of T cells with direct allospecificity and how self-restricted T cells recognise allogeneic MHC-peptide complexes. These theories, both of which are supported by functional and structural data, suggest that T cells recognising allogeneic MHC-peptide complexes focus either on the allopeptides bound to the allo-MHC molecules or the allo-MHC molecules themselves. We discuss how direct alloimmune responses may be sustained long term, the consequences of this for graft outcome and highlight novel strategies which are currently being investigated as a potential means of reducing rejection mediated through this pathway.
Collapse
Affiliation(s)
- Dominic A Boardman
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, SE1 9RT UK ; NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, SE1 9RT UK
| | - Jacinta Jacob
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, SE1 9RT UK
| | - Lesley A Smyth
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, SE1 9RT UK ; School of Health, Sport and Bioscience, Stratford Campus, University of East London, London, E15 4LZ UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, SE1 9RT UK ; NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, SE1 9RT UK
| | - Robert I Lechler
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, SE1 9RT UK ; NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, SE1 9RT UK
| |
Collapse
|
166
|
Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, Herold KC, Lares A, Lee MR, Li K, Liu W, Long SA, Masiello LM, Nguyen V, Putnam AL, Rieck M, Sayre PH, Tang Q. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2016; 7:315ra189. [PMID: 26606968 DOI: 10.1126/scitranslmed.aad4134] [Citation(s) in RCA: 789] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that occurs in genetically susceptible individuals. Regulatory T cells (Tregs) have been shown to be defective in the autoimmune disease setting. Thus, efforts to repair or replace Tregs in T1D may reverse autoimmunity and protect the remaining insulin-producing β cells. On the basis of this premise, a robust technique has been developed to isolate and expand Tregs from patients with T1D. The expanded Tregs retained their T cell receptor diversity and demonstrated enhanced functional activity. We report on a phase 1 trial to assess safety of Treg adoptive immunotherapy in T1D. Fourteen adult subjects with T1D, in four dosing cohorts, received ex vivo-expanded autologous CD4(+)CD127(lo/-)CD25(+) polyclonal Tregs (0.05 × 10(8) to 26 × 10(8) cells). A subset of the adoptively transferred Tregs was long-lived, with up to 25% of the peak level remaining in the circulation at 1 year after transfer. Immune studies showed transient increases in Tregs in recipients and retained a broad Treg FOXP3(+)CD4(+)CD25(hi)CD127(lo) phenotype long-term. There were no infusion reactions or cell therapy-related high-grade adverse events. C-peptide levels persisted out to 2+ years after transfer in several individuals. These results support the development of a phase 2 trial to test efficacy of the Treg therapy.
Collapse
Affiliation(s)
- Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Mark Fitch
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Stephen E Gitelman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shipra Gupta
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Marc K Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Angela Lares
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael R Lee
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kelvin Li
- KineMed Inc., Emeryville, CA 94608, USA
| | - Weihong Liu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - S Alice Long
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Lisa M Masiello
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vinh Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amy L Putnam
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mary Rieck
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Peter H Sayre
- Division of Hematology-Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
167
|
Bar-Or A, Steinman L, Behne JM, Benitez-Ribas D, Chin PS, Clare-Salzler M, Healey D, Kim JI, Kranz DM, Lutterotti A, Martin R, Schippling S, Villoslada P, Wei CH, Weiner HL, Zamvil SS, Smith TJ, Yeaman MR. Restoring immune tolerance in neuromyelitis optica: Part II. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e277. [PMID: 27648464 PMCID: PMC5015540 DOI: 10.1212/nxi.0000000000000277] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 07/15/2016] [Indexed: 12/22/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMO/SD) and its clinical variants have at their core the loss of immune tolerance to aquaporin-4 and perhaps other autoantigens. The characteristic phenotype is disruption of astrocyte function and demyelination of spinal cord, optic nerves, and particular brain regions. In this second of a 2-part article, we present further perspectives regarding the pathogenesis of NMO/SD and how this disease might be amenable to emerging technologies aimed at restoring immune tolerance to disease-implicated self-antigens. NMO/SD appears to be particularly well-suited for these strategies since aquaporin-4 has already been identified as the dominant autoantigen. The recent technical advances in reintroducing immune tolerance in experimental models of disease as well as in humans should encourage quantum leaps in this area that may prove productive for novel therapy. In this part of the article series, the potential for regulatory T and B cells is brought into focus, as are new approaches to oral tolerization. Finally, a roadmap is provided to help identify potential issues in clinical development and guide applications in tolerization therapy to solving NMO/SD through the use of emerging technologies. Each of these perspectives is intended to shine new light on potential cures for NMO/SD and other autoimmune diseases, while sparing normal host defense mechanisms.
Collapse
Affiliation(s)
- Amit Bar-Or
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Larry Steinman
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Jacinta M Behne
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Daniel Benitez-Ribas
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Peter S Chin
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Michael Clare-Salzler
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Donald Healey
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - James I Kim
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - David M Kranz
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Andreas Lutterotti
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Roland Martin
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Sven Schippling
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Pablo Villoslada
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Cheng-Hong Wei
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Howard L Weiner
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Scott S Zamvil
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Terry J Smith
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Michael R Yeaman
- Neuroimmunology Unit and Experimental Therapeutics Program (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada; Department of Neurology (L.S.), Stanford University School of Medicine, Palo Alto; The Guthy-Jackson Charitable Foundation (J.M.B.), San Diego, CA; Department of Gastroenterology (D.B.-R., P.V.), Hospital Clínic, CIBERehd and Center of Neuroimmunology & Inflammatory Bowel Disease, Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain; Genentech, Inc. (P.S.C.), South San Francisco, CA; Department of Pathology (M.C.-S.), University of Florida School of Medicine, Gainesville; Opexa Therapeutics (D.H.), The Woodlands, TX; Department of Surgery (J.I.K.), Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, MA; Department of Biochemistry (D.M.K.), University of Illinois, Urbana; Neuroimmunology and MS Research (A.L., R.M., S.S.), Department of Neurology, University Hospital Zurich, University Zurich, Switzerland; Ann Romney Center for Neurologic Diseases (H.L.W.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Neurology and Program in Immunology (S.S.Z.), University of California, San Francisco School of Medicine; Department of Ophthalmology and Visual Sciences (T.J.S.), Kellogg Eye Center, and Division of Metabolism, Endocrine and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine & Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles; and Harbor-UCLA Medical Center & LABioMed at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| |
Collapse
|
168
|
Kordasti S, Costantini B, Seidl T, Perez Abellan P, Martinez Llordella M, McLornan D, Diggins KE, Kulasekararaj A, Benfatto C, Feng X, Smith A, Mian SA, Melchiotti R, de Rinaldis E, Ellis R, Petrov N, Povoleri GAM, Chung SS, Thomas NSB, Farzaneh F, Irish JM, Heck S, Young NS, Marsh JCW, Mufti GJ. Deep phenotyping of Tregs identifies an immune signature for idiopathic aplastic anemia and predicts response to treatment. Blood 2016; 128:1193-205. [PMID: 27281795 PMCID: PMC5009512 DOI: 10.1182/blood-2016-03-703702] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/01/2016] [Indexed: 11/20/2022] Open
Abstract
Idiopathic aplastic anemia (AA) is an immune-mediated and serious form of bone marrow failure. Akin to other autoimmune diseases, we have previously shown that in AA regulatory T cells (Tregs) are reduced in number and function. The aim of this study was to further characterize Treg subpopulations in AA and investigate the potential correlation between specific Treg subsets and response to immunosuppressive therapy (IST) as well as their in vitro expandability for potential clinical use. Using mass cytometry and an unbiased multidimensional analytical approach, we identified 2 specific human Treg subpopulations (Treg A and Treg B) with distinct phenotypes, gene expression, expandability, and function. Treg B predominates in IST responder patients, has a memory/activated phenotype (with higher expression of CD95, CCR4, and CD45RO within FOXP3(hi), CD127(lo) Tregs), expresses the interleukin-2 (IL-2)/STAT5 pathway and cell-cycle commitment genes. Furthermore, in vitro-expanded Tregs become functional and take on the characteristics of Treg B. Collectively, this study identifies human Treg subpopulations that can be used as predictive biomarkers for response to IST in AA and potentially other autoimmune diseases. We also show that Tregs from AA patients are IL-2-sensitive and expandable in vitro, suggesting novel therapeutic approaches such as low-dose IL-2 therapy and/or expanded autologous Tregs and meriting further exploration.
Collapse
Affiliation(s)
- Shahram Kordasti
- Department of Haematological Medicine, King's College London, London, United Kingdom; Haematological Medicine, King's College Hospital, London, United Kingdom
| | - Benedetta Costantini
- Department of Haematological Medicine, King's College London, London, United Kingdom; Haematological Medicine, King's College Hospital, London, United Kingdom
| | - Thomas Seidl
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | | | - Marc Martinez Llordella
- Division of Transplantation Immunology & Mucosal Biology, King's College London, London, United Kingdom
| | - Donal McLornan
- Haematological Medicine, King's College Hospital, London, United Kingdom
| | | | | | - Cinzia Benfatto
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and
| | - Alexander Smith
- Department of Haematological Medicine, King's College London, London, United Kingdom; Haematological Medicine, King's College Hospital, London, United Kingdom
| | - Syed A Mian
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Rossella Melchiotti
- National Institute for Health Research Biomedical Research Centre, King's College London, London, United Kingdom
| | - Emanuele de Rinaldis
- National Institute for Health Research Biomedical Research Centre, King's College London, London, United Kingdom
| | - Richard Ellis
- National Institute for Health Research Biomedical Research Centre, King's College London, London, United Kingdom
| | - Nedyalko Petrov
- National Institute for Health Research Biomedical Research Centre, King's College London, London, United Kingdom
| | - Giovanni A M Povoleri
- Division of Transplantation Immunology & Mucosal Biology, King's College London, London, United Kingdom
| | - Sun Sook Chung
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | - N Shaun B Thomas
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Farzin Farzaneh
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Jonathan M Irish
- Department of Cancer Biology, Vanderbilt University, Nashville, TN
| | - Susanne Heck
- National Institute for Health Research Biomedical Research Centre, King's College London, London, United Kingdom
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD; and
| | - Judith C W Marsh
- Department of Haematological Medicine, King's College London, London, United Kingdom; Haematological Medicine, King's College Hospital, London, United Kingdom
| | - Ghulam J Mufti
- Department of Haematological Medicine, King's College London, London, United Kingdom; Haematological Medicine, King's College Hospital, London, United Kingdom
| |
Collapse
|
169
|
Hu M, Wang YM, Wang Y, Zhang GY, Zheng G, Yi S, O'Connell PJ, Harris DCH, Alexander SI. Regulatory T cells in kidney disease and transplantation. Kidney Int 2016; 90:502-514. [PMID: 27263492 DOI: 10.1016/j.kint.2016.03.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/06/2016] [Accepted: 03/17/2016] [Indexed: 01/03/2023]
Abstract
Regulatory T cells (Tregs) have been shown to be important in maintaining immune homeostasis and preventing autoimmune disease, including autoimmune kidney disease. It is also likely that they play a role in limiting kidney transplant rejection and potentially in promoting transplant tolerance. Although other subsets of Tregs exist, the most potent and well-defined Tregs are the Foxp3 expressing CD4(+) Tregs derived from the thymus or generated peripherally. These CD4(+)Foxp3(+) Tregs limit autoimmune renal disease in animal models, especially chronic kidney disease, and kidney transplantation. Furthermore, other subsets of Tregs, including CD8 Tregs, may play a role in immunosuppression in kidney disease. The development and protective mechanisms of Tregs in kidney disease and kidney transplantation involve multiple mechanisms of suppression. Here we review the development and function of CD4(+)Foxp3(+) Tregs. We discuss the specific application of Tregs as a therapeutic strategy to prevent kidney disease and to limit kidney transplant rejection and detail clinical trials in this area of transplantation.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia; Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Geoff Y Zhang
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Shounan Yi
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Philip J O'Connell
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - David C H Harris
- Centre for Transplantation and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, The Children's Hospital at Westmead, University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
170
|
Chen Y, Jeffery HC, Hunter S, Bhogal R, Birtwistle J, Braitch MK, Roberts S, Ming M, Hannah J, Thomas C, Adali G, Hübscher SG, Syn W, Afford S, Lalor PF, Adams DH, Oo YH. Human intrahepatic regulatory T cells are functional, require IL-2 from effector cells for survival, and are susceptible to Fas ligand-mediated apoptosis. Hepatology 2016; 64:138-50. [PMID: 26928938 PMCID: PMC4950043 DOI: 10.1002/hep.28517] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 01/25/2016] [Accepted: 02/25/2016] [Indexed: 01/02/2023]
Abstract
UNLABELLED Regulatory T cells (Treg ) suppress T effector cell proliferation and maintain immune homeostasis. Autoimmune liver diseases persist despite high frequencies of Treg in the liver, suggesting that the local hepatic microenvironment might affect Treg stability, survival, and function. We hypothesized that interactions between Treg and endothelial cells during recruitment and then with epithelial cells within the liver affect Treg stability, survival, and function. To model this, we explored the function of Treg after migration through human hepatic sinusoidal-endothelium (postendothelial migrated Treg [PEM Treg ]) and the effect of subsequent interactions with cholangiocytes and local proinflammatory cytokines on survival and stability of Treg . Our findings suggest that the intrahepatic microenvironment is highly enriched with proinflammatory cytokines but deficient in the Treg survival cytokine interleukin (IL)-2. Migration through endothelium into a model mimicking the inflamed liver microenvironment did not affect Treg stability; however, functional capacity was reduced. Furthermore, the addition of exogenous IL-2 enhanced PEM Treg phosphorylated STAT5 signaling compared with PEMCD8. CD4 and CD8 T cells are the main source of IL-2 in the inflamed liver. Liver-infiltrating Treg reside close to bile ducts and coculture with cholangiocytes or their supernatants induced preferential apoptosis of Treg compared with CD8 effector cells. Treg from diseased livers expressed high levels of CD95, and their apoptosis was inhibited by IL-2 or blockade of CD95. CONCLUSION Recruitment through endothelium does not impair Treg stability, but a proinflammatory microenvironment deficient in IL-2 leads to impaired function and increased susceptibility of Treg to epithelial cell-induced Fas-mediated apoptosis. These results provide a mechanism to explain Treg dysfunction in inflamed tissues and suggest that IL-2 supplementation, particularly if used in conjunction with Treg therapy, could restore immune homeostasis in inflammatory and autoimmune liver disease. (Hepatology 2016;64:138-150).
Collapse
Affiliation(s)
- Yung‐Yi Chen
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom,Institute of Immunology and ImmunotherapyUniversity of Birmingham, BirminghamUnited Kingdom
| | - Hannah C. Jeffery
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom,Institute of Immunology and ImmunotherapyUniversity of Birmingham, BirminghamUnited Kingdom
| | - Stuart Hunter
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Ricky Bhogal
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Jane Birtwistle
- Clinical Immunology DepartmentUniversity Hospital Birmingham NHS Foundation TrustBirminghamUnited Kingdom
| | - Manjit Kaur Braitch
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Sheree Roberts
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Mikaela Ming
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Jack Hannah
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Clare Thomas
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Gupse Adali
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom
| | - Stefan G. Hübscher
- Department of Cellular PathologyQueen Elizabeth Hospital BirminghamUnited Kingdom
| | - Wing‐Kin Syn
- The Institute of HepatologyLondonUnited Kingdom,Division of Gastroenterology and HepatologyThe Medical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Simon Afford
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom,Institute of Immunology and ImmunotherapyUniversity of Birmingham, BirminghamUnited Kingdom
| | - Patricia F. Lalor
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom,Institute of Immunology and ImmunotherapyUniversity of Birmingham, BirminghamUnited Kingdom
| | - David H. Adams
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom,Institute of Immunology and ImmunotherapyUniversity of Birmingham, BirminghamUnited Kingdom
| | - Ye H. Oo
- Centre for Liver Research and NIHR Birmingham Liver Biomedical Research UnitUniversity of BirminghamBirminghamUnited Kingdom,Institute of Immunology and ImmunotherapyUniversity of Birmingham, BirminghamUnited Kingdom
| |
Collapse
|
171
|
Ezzelarab MB, Zhang H, Guo H, Lu L, Zahorchak AF, Wiseman RW, Nalesnik MA, Bhama JK, Cooper DKC, Thomson AW. Regulatory T Cell Infusion Can Enhance Memory T Cell and Alloantibody Responses in Lymphodepleted Nonhuman Primate Heart Allograft Recipients. Am J Transplant 2016; 16:1999-2015. [PMID: 26700196 PMCID: PMC4919255 DOI: 10.1111/ajt.13685] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/25/2015] [Accepted: 12/13/2015] [Indexed: 01/25/2023]
Abstract
The ability of regulatory T cells (Treg) to prolong allograft survival and promote transplant tolerance in lymphodepleted rodents is well established. Few studies, however, have addressed the therapeutic potential of adoptively transferred, CD4(+) CD25(+) CD127(-) Foxp3(+) (Treg) in clinically relevant large animal models. We infused ex vivo-expanded, functionally stable, nonselected Treg (up to a maximum cumulative dose of 1.87 billion cells) into antithymocyte globulin-lymphodepleted, MHC-mismatched cynomolgus monkey heart graft recipients before homeostatic recovery of effector T cells. The monkeys also received tacrolimus, anti-interleukin-6 receptor monoclonal antibodies and tapered rapamycin maintenance therapy. Treg administration in single or multiple doses during the early postsurgical period (up to 1 month posttransplantation), when host T cells were profoundly depleted, resulted in inferior graft function compared with controls. This was accompanied by increased incidences of effector memory T cells, enhanced interferon-γ production by host CD8(+) T cells, elevated levels of proinflammatory cytokines, and antidonor alloantibodies. The findings caution against infusion of Treg during the early posttransplantation period after lymphodepletion. Despite marked but transient increases in Treg relative to endogenous effector T cells and use of reputed "Treg-friendly" agents, the host environment/immune effector mechanisms instigated under these conditions can perturb rather than favor the potential therapeutic efficacy of adoptively transferred Treg.
Collapse
Affiliation(s)
- M. B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - H. Zhang
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - H. Guo
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - A. F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine
| | - R. W. Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI
| | - M. A. Nalesnik
- Department of Pathology, University of Pittsburgh School of Medicine
| | - J. K. Bhama
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine
| | - D. K. C. Cooper
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine
| | - A. W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine,Department of Immunology, University of Pittsburgh School of Medicine,Corresponding author: Angus W. Thomson PhD DSc,
| |
Collapse
|
172
|
Abstract
PURPOSE OF REVIEW Transformative medical and surgical advances have remarkably improved short-term survival after liver transplantation. There is, however, pervasive concern that the cumulative toxicities of modern immunosuppression regimens severely compromise both quality and quantity of life for liver transplant recipients. The inherently tolerogenic nature of the liver offers the tantalizing opportunity to change the current paradigm of nonspecific and lifelong immunosuppression. Safe minimization or discontinuation of immunosuppression without damage to the liver allograft is an attractive strategy to improve long-term survival after liver transplantation. RECENT FINDINGS Recent prospective, multicenter clinical trials have demonstrated that immunosuppression can be safely withdrawn from selected liver transplant recipients with preservation of allograft histology. These successes have spurred multiple avenues of investigation to identify peripheral blood and/or tissue biomarkers and delineate mechanisms of tolerance. Concomitant advances in the ability to expand regulatory T cells in the laboratory have spawned clinical trials to facilitate immunosuppression minimization and/or discontinuation. SUMMARY This review will delineate the unique liver immunobiology that has driven the recent clinical trials to unmask spontaneous tolerance or induce tolerance for liver transplant recipients. The emerging results of these trials over the next 5 years hold promise to reduce the burden of lifelong immunosuppression and thereby optimize the long-term health of liver transplant recipients.
Collapse
|
173
|
Hoeppli RE, MacDonald KG, Levings MK, Cook L. How antigen specificity directs regulatory T-cell function: self, foreign and engineered specificity. HLA 2016; 88:3-13. [PMID: 27256587 DOI: 10.1111/tan.12822] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 04/29/2016] [Indexed: 02/06/2023]
Abstract
Regulatory T cells (Tregs) are a suppressive subset of T cells that have important roles in maintaining self-tolerance and preventing immunopathology. The T-cell receptor (TCR) and its antigen specificity play a dominant role in the differentiation of cells to a Treg fate, either in the thymus or in the periphery. This review focuses on the effects of the TCR and its antigen specificity on Treg biology. The role of Tregs with specificity for self-antigen has primarily been studied in the context of autoimmune disease, although recent studies have focused on their role in steady-state conditions. The role of Tregs that are specific for pathogens, dietary antigens and allergens is much less studied, although recent data suggest a significant and previously underappreciated role for Tregs during memory responses to a wide range of foreign antigens. The development of TCR- or chimeric antigen receptor (CAR)-transduced T cells means we are now able to engineer Tregs with disease-relevant antigen specificities, paving the way for ensuring specificity with Treg-based therapies. Understanding the role that antigens play in driving the generation and function of Tregs is critical for defining the pathophysiology of many immune-mediated diseases, and developing new therapeutic interventions.
Collapse
Affiliation(s)
- R E Hoeppli
- Department of Surgery, University of British Columbia and Child & Family Research Institute, Vancouver, Canada
| | - K G MacDonald
- Department of Surgery, University of British Columbia and Child & Family Research Institute, Vancouver, Canada
| | - M K Levings
- Department of Surgery, University of British Columbia and Child & Family Research Institute, Vancouver, Canada
| | - L Cook
- Department of Surgery, University of British Columbia and Child & Family Research Institute, Vancouver, Canada
| |
Collapse
|
174
|
Antigen-specificity using chimeric antigen receptors: the future of regulatory T-cell therapy? Biochem Soc Trans 2016; 44:342-8. [DOI: 10.1042/bst20150247] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 12/24/2022]
Abstract
Adoptive regulatory T-cell (Treg) therapy using autologous Tregs expanded ex vivo is a promising therapeutic approach which is currently being investigated clinically as a means of treating various autoimmune diseases and transplant rejection. Despite this, early results have highlighted the need for potent Tregs to yield a substantial clinical advantage. One way to achieve this is to create antigen-specific Tregs which have been shown in pre-clinical animal models to have an increased potency at suppressing undesired immune responses, compared to polyclonal Tregs. This mini review outlines where Treg therapy currently stands and discusses the approaches which may be taken to generate antigen-specific Tregs, including the potential use of chimeric antigen receptors (CARs), for future clinical trials.
Collapse
|
175
|
Canavan JB, Scottà C, Vossenkämper A, Goldberg R, Elder MJ, Shoval I, Marks E, Stolarczyk E, Lo JW, Powell N, Fazekasova H, Irving PM, Sanderson JD, Howard JK, Yagel S, Afzali B, MacDonald TT, Hernandez-Fuentes MP, Shpigel NY, Lombardi G, Lord GM. Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy for Crohn's disease. Gut 2016; 65:584-94. [PMID: 25715355 PMCID: PMC4819603 DOI: 10.1136/gutjnl-2014-306919] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 12/23/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Thymus-derived regulatory T cells (Tregs) mediate dominant peripheral tolerance and treat experimental colitis. Tregs can be expanded from patient blood and were safely used in recent phase 1 studies in graft versus host disease and type 1 diabetes. Treg cell therapy is also conceptually attractive for Crohn's disease (CD). However, barriers exist to this approach. The stability of Tregs expanded from Crohn's blood is unknown. The potential for adoptively transferred Tregs to express interleukin-17 and exacerbate Crohn's lesions is of concern. Mucosal T cells are resistant to Treg-mediated suppression in active CD. The capacity for expanded Tregs to home to gut and lymphoid tissue is unknown. METHODS To define the optimum population for Treg cell therapy in CD, CD4(+)CD25(+)CD127(lo)CD45RA(+) and CD4(+)CD25(+)CD127(lo)CD45RA(-) Treg subsets were isolated from patients' blood and expanded in vitro using a workflow that can be readily transferred to a good manufacturing practice background. RESULTS Tregs can be expanded from the blood of patients with CD to potential target dose within 22-24 days. Expanded CD45RA(+) Tregs have an epigenetically stable FOXP3 locus and do not convert to a Th17 phenotype in vitro, in contrast to CD45RA(-) Tregs. CD45RA(+) Tregs highly express α4β7 integrin, CD62L and CC motif receptor 7 (CCR7). CD45RA(+) Tregs also home to human small bowel in a C.B-17 severe combined immune deficiency (SCID) xenotransplant model. Importantly, in vitro expansion enhances the suppressive ability of CD45RA(+) Tregs. These cells also suppress activation of lamina propria and mesenteric lymph node lymphocytes isolated from inflamed Crohn's mucosa. CONCLUSIONS CD4(+)CD25(+)CD127(lo)CD45RA(+) Tregs may be the most appropriate population from which to expand Tregs for autologous Treg therapy for CD, paving the way for future clinical trials.
Collapse
Affiliation(s)
- James B Canavan
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Gastroenterology, Guy's & St Thomas’ NHS Foundation Trust, London, UK
| | - Cristiano Scottà
- Medical Research Council Centre for Transplantation, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Immunoregulation and Immune Intervention, King's College London, London, UK
| | - Anna Vossenkämper
- Blizard Institute, Barts and the London School of Medicine and Dentistry, London, UK
| | - Rimma Goldberg
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Gastroenterology, Guy's & St Thomas’ NHS Foundation Trust, London, UK
| | - Matthew J Elder
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK
| | - Irit Shoval
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Ellen Marks
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK
| | - Emilie Stolarczyk
- National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Jonathan W Lo
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK
| | - Nick Powell
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Gastroenterology, Guy's & St Thomas’ NHS Foundation Trust, London, UK
| | - Henrieta Fazekasova
- Medical Research Council Centre for Transplantation, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Immunoregulation and Immune Intervention, King's College London, London, UK
| | - Peter M Irving
- Department of Gastroenterology, Guy's & St Thomas’ NHS Foundation Trust, London, UK
| | - Jeremy D Sanderson
- Department of Gastroenterology, Guy's & St Thomas’ NHS Foundation Trust, London, UK
| | - Jane K Howard
- National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Division of Diabetes and Nutritional Sciences, King's College London, London, UK
| | - Simcha Yagel
- Department of Obstetrics & Gynaecology, Hadassah University Hospital, Jerusalem, Israel
| | - Behdad Afzali
- Medical Research Council Centre for Transplantation, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Immunoregulation and Immune Intervention, King's College London, London, UK
| | - Thomas T MacDonald
- Blizard Institute, Barts and the London School of Medicine and Dentistry, London, UK
| | - Maria P Hernandez-Fuentes
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK
| | - Nahum Y Shpigel
- The Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Giovanna Lombardi
- Medical Research Council Centre for Transplantation, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK,Department of Immunoregulation and Immune Intervention, King's College London, London, UK
| | - Graham M Lord
- Medical Research Council Centre for Transplantation, King's College London, London, UK,Department of Experimental Immunobiology, King's College London, London, UK,National Institute for Health Research Biomedical Research Centre at Guy's and St. Thomas’ NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
176
|
MacDonald KG, Hoeppli RE, Huang Q, Gillies J, Luciani DS, Orban PC, Broady R, Levings MK. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor. J Clin Invest 2016; 126:1413-24. [PMID: 26999600 DOI: 10.1172/jci82771] [Citation(s) in RCA: 373] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 02/04/2016] [Indexed: 12/21/2022] Open
Abstract
Adoptive immunotherapy with regulatory T cells (Tregs) is a promising treatment for allograft rejection and graft-versus-host disease (GVHD). Emerging data indicate that, compared with polyclonal Tregs, disease-relevant antigen-specific Tregs may have numerous advantages, such as a need for fewer cells and reduced risk of nonspecific immune suppression. Current methods to generate alloantigen-specific Tregs rely on expansion with allogeneic antigen-presenting cells, which requires access to donor and recipient cells and multiple MHC mismatches. The successful use of chimeric antigen receptors (CARs) for the generation of antigen-specific effector T cells suggests that a similar approach could be used to generate alloantigen-specific Tregs. Here, we have described the creation of an HLA-A2-specific CAR (A2-CAR) and its application in the generation of alloantigen-specific human Tregs. In vitro, A2-CAR-expressing Tregs maintained their expected phenotype and suppressive function before, during, and after A2-CAR-mediated stimulation. In mouse models, human A2-CAR-expressing Tregs were superior to Tregs expressing an irrelevant CAR at preventing xenogeneic GVHD caused by HLA-A2+ T cells. Together, our results demonstrate that use of CAR technology to generate potent, functional, and stable alloantigen-specific human Tregs markedly enhances their therapeutic potential in transplantation and sets the stage for using this approach for making antigen-specific Tregs for therapy of multiple diseases.
Collapse
|
177
|
Kenney LL, Shultz LD, Greiner DL, Brehm MA. Humanized Mouse Models for Transplant Immunology. Am J Transplant 2016; 16:389-97. [PMID: 26588186 PMCID: PMC5283075 DOI: 10.1111/ajt.13520] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 01/25/2023]
Abstract
Our understanding of the molecular pathways that control immune responses, particularly immunomodulatory molecules that control the extent and duration of an immune response, have led to new approaches in the field of transplantation immunology to induce allograft survival. These molecular pathways are being defined precisely in murine models and translated into clinical practice; however, many of the newly available drugs are human-specific reagents. Furthermore, many species-specific differences exist between mouse and human immune systems. Recent advances in the development of humanized mice, namely, immunodeficient mice engrafted with functional human immune systems, have led to the availability of a small animal model for the study of human immune responses. Humanized mice represent an important preclinical model system for evaluation of new drugs and identification of the mechanisms underlying human allograft rejection without putting patients at risk. This review highlights recent advances in the development of humanized mice and their use as preclinical models for the study of human allograft responses.
Collapse
Affiliation(s)
- Laurie L Kenney
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| | | | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605,Corresponding Author: Dale L. Greiner, PhD, University of Massachusetts Medical School, 368 Plantation Street, AS7-2051, Worcester, MA 01605, Office: 508-856-1911, Fax: 508-856-4093,
| | - Michael A. Brehm
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605
| |
Collapse
|
178
|
Safinia N, Becker PD, Vaikunthanathan T, Xiao F, Lechler R, Lombardi G. Humanized Mice as Preclinical Models in Transplantation. Methods Mol Biol 2016; 1371:177-196. [PMID: 26530801 DOI: 10.1007/978-1-4939-3139-2_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Animal models have been instrumental in our understanding of the mechanisms of rejection and the testing of novel treatment options in the context of transplantation. We have now entered an exciting era with research on humanized mice driving advances in translational studies and in our understanding of the function of human cells in response to pathogens and cancer as well as the recognition of human allogeneic tissues in vivo. In this chapter we provide a historical overview of humanized mouse models of transplantation to date, outlining the distinct strains and share our experiences in the study of human transplantation immunology.
Collapse
Affiliation(s)
- N Safinia
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - P D Becker
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - T Vaikunthanathan
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - F Xiao
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - R Lechler
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK
| | - G Lombardi
- MRC Centre for Transplantation, King's College London, Guy's Hospital, 5th Floor Tower Wing, London, SE1 9RT, UK.
| |
Collapse
|
179
|
Autoimmune Hepatitis: Progress from Global Immunosuppression to Personalised Regulatory T Cell Therapy. Can J Gastroenterol Hepatol 2016; 2016:7181685. [PMID: 27446862 PMCID: PMC4904688 DOI: 10.1155/2016/7181685] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 11/20/2015] [Indexed: 12/23/2022] Open
Abstract
Autoimmune hepatitis (AIH) is an immune mediated liver injury. The precise aetiology of AIH is still unknown but current evidence suggests both genetic and environmental factors are involved. Breakdown in peripheral self-tolerance, and impaired functions of FOXP3(+) regulatory T cell along with effector cell resistance to suppression at the tissue level seem to play an important role in AIH immunopathogenesis. AIH is predominantly a T lymphocytes driven disease but B lymphocytes are also involved in the immunopathology. Innate immune cells are crucial in the initial onset of disease and their response is followed by adaptive T (Th1, Th17, and cytotoxic T cells) and B cell responses evidenced by liver histology and peripheral blood serology. Standard treatment regimens involving steroid and immunosuppressive medications lead to global immune suppression requiring life-long therapy with many side effects. Biologic therapies have been attempted but duration of remission is short-lived. Future direction of diagnosis and treatment for AIH should be guided by "omics" and the immunology profile of the individual patient and clinicians should aim to deliver personalised medicine for their patients. Cell therapy such as infusion of autologous, antigen-specific, and liver-homing regulatory T cells to restore hepatic immune tolerance may soon be a potential future treatment for AIH patients.
Collapse
|
180
|
Swann JW, Garden OA. Novel immunotherapies for immune-mediated haemolytic anaemia in dogs and people. Vet J 2016; 207:13-9. [DOI: 10.1016/j.tvjl.2015.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
181
|
Hahn SA, Bellinghausen I, Trinschek B, Becker C. Translating Treg Therapy in Humanized Mice. Front Immunol 2015; 6:623. [PMID: 26697017 PMCID: PMC4677486 DOI: 10.3389/fimmu.2015.00623] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/30/2022] Open
Abstract
Regulatory T cells (Treg) control immune cell function as well as non-immunological processes. Their far-reaching regulatory activities suggest their functional manipulation as a means to sustainably and causally intervene with the course of diseases. Preclinical tools and strategies are however needed to further test and develop interventional strategies outside the human body. “Humanized” mouse models consisting of mice engrafted with human immune cells and tissues provide new tools to analyze human Treg ontogeny, immunobiology, and therapy. Here, we summarize the current state of humanized mouse models as a means to study human Treg function at the molecular level and to design strategies to harness these cells for therapeutic purposes.
Collapse
Affiliation(s)
- Susanne A Hahn
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Iris Bellinghausen
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Bettina Trinschek
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Christian Becker
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| |
Collapse
|
182
|
Chera M, Hamel Y, Baillou C, Touil S, Guillot-Delost M, Charlotte F, Kossir L, Simonin G, Maury S, Cohen JL, Lemoine FM. Generation of Human Alloantigen-Specific Regulatory T Cells under Good Manufacturing Practice-Compliant Conditions for Cell Therapy. Cell Transplant 2015; 24:2527-40. [DOI: 10.3727/096368914x683566] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Natural regulatory T cells (Tregs) may have a great therapeutic potential to induce tolerance in allogeneic cells and organ transplantations. In mice, we showed that alloantigen-specific Tregs (spe-Tregs) were more efficient than polyclonal Tregs (poly-Tregs) in controlling graft-versus-host disease (GVHD). Here we describe a clinical-grade compliant method for generating human spe-Tregs. Tregs were enriched from leukapheresis products with anti-CD25 immunomagnetic beads, primed twice by allogeneic mature monocyte-derived dendritic cells (mDCs), and cultured during 3 weeks in medium containing interleukin 2 (IL-2), IL-15, and rapamycin. After 3 weeks of culture, final cell products were expanded 8.3-fold from the initial CD25+ purifications. Immunophenotypic analyses of final cells indicate that they were composed of 88 ± 2.6% of CD4+ T cells, all expressing Treg-specific markers (FOXP3, Helios, GARP, LAP, and CD152). Spe-Tregs were highly suppressive in vitro and also in vivo using a xeno-GVHD model established in immunodeficient mice. The specificity of their suppressive activity was demonstrated on their ability to significantly suppress the proliferation of autologous effector T cells stimulated by the same mDCs compared to third-party mDCs. Our data provide evidence that functional alloantigen Tregs can be generated under clinical-grade compliant conditions. Taking into account that 130 × 106 CD25+ cells can be obtained at large scale from standard leukapheresis, our cell process may give rise to a theoretical final number of 1 × 109 spe-Tregs. Thus, using our strategy, we can propose to prepare spe-Tregs for clinical trials designed to control HLA-mismatched GVHD or organ transplantation rejection.
Collapse
Affiliation(s)
- Mustapha Chera
- AP-HP, University Hospital La Pitié-Salpêtrière, Department of Biotherapies, Paris, France
- Center of Clinical Investigation in Biotherapies 1420, University Hospital La Pitié-Salpêtrière, Paris, France
| | - Yamina Hamel
- Sorbonne Universités, UPMC Univ Paris 06, UMR-S CR7, CIMI-Paris, Paris, France
| | - Claude Baillou
- Sorbonne Universités, UPMC Univ Paris 06, UMR-S CR7, CIMI-Paris, Paris, France
- INSERM, UMR S 1135, CIMI-Paris, Paris, France
| | - Soumia Touil
- CNRS, UMR 7211, Immunology Immunopathology and Immunotherapy, Paris, France
| | - Maude Guillot-Delost
- Sorbonne Universités, UPMC Univ Paris 06, UMR-S CR7, CIMI-Paris, Paris, France
- INSERM, UMR S 1135, CIMI-Paris, Paris, France
| | - Frédéric Charlotte
- AP-HP, University Hospital La Pitié-Salpêtrière, Department of Pathology, Paris, France
| | - Laila Kossir
- AP-HP, University Hospital La Pitié-Salpêtrière, Department of Biotherapies, Paris, France
- Center of Clinical Investigation in Biotherapies 1420, University Hospital La Pitié-Salpêtrière, Paris, France
| | - Ghislaine Simonin
- AP-HP, University Hospital La Pitié-Salpêtrière, Department of Biotherapies, Paris, France
- Center of Clinical Investigation in Biotherapies 1420, University Hospital La Pitié-Salpêtrière, Paris, France
| | - Sébastien Maury
- AP-HP, Henri-Mondor Hospital, Department of Clinical Hematology, Créteil, France
- Université Paris-Est, UMR-S955, UPEC, Créteil, France
- INSERM, U955, Team 21, Créteil, France
| | - José L. Cohen
- Université Paris-Est, UMR-S955, UPEC, Créteil, France
- INSERM, U955, Team 21, Créteil, France
- AP-HP, Henri-Mondor - A. Chenevier Hospital, CIC-BT-504, Créteil, France
| | - François M. Lemoine
- AP-HP, University Hospital La Pitié-Salpêtrière, Department of Biotherapies, Paris, France
- Center of Clinical Investigation in Biotherapies 1420, University Hospital La Pitié-Salpêtrière, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR-S CR7, CIMI-Paris, Paris, France
- INSERM, UMR S 1135, CIMI-Paris, Paris, France
| |
Collapse
|
183
|
Spence A, Klementowicz JE, Bluestone JA, Tang Q. Targeting Treg signaling for the treatment of autoimmune diseases. Curr Opin Immunol 2015; 37:11-20. [PMID: 26432763 PMCID: PMC4679451 DOI: 10.1016/j.coi.2015.09.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/30/2015] [Accepted: 09/10/2015] [Indexed: 12/16/2022]
Abstract
Regulatory T (Treg) cells are crucial players in the prevention of autoimmunity. Treg lineage commitment and functional stability are influenced by selected extracellular signals from the local environment, shaped by distinctive intracellular signaling network, and secured by their unique epigenetic profile. Recent advances in our understanding of the complex processes of Treg lineage differentiation, maintenance, and function has paved the way for developing strategies to manipulate these important cells for therapeutic benefit in many diseases. In this review, we will summarize recent advances in our understanding of Treg biology as well as Treg-targeted therapies in the context of autoimmune disease.
Collapse
Affiliation(s)
- Allyson Spence
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joanna E Klementowicz
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeffrey A Bluestone
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
184
|
|
185
|
Gregori S, Passerini L, Roncarolo MG. Clinical Outlook for Type-1 and FOXP3(+) T Regulatory Cell-Based Therapy. Front Immunol 2015; 6:593. [PMID: 26635807 PMCID: PMC4658444 DOI: 10.3389/fimmu.2015.00593] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/05/2015] [Indexed: 12/18/2022] Open
Abstract
T regulatory cells (Tregs) are subsets of T lymphocytes specialized in modulating antigen-specific immune responses in vivo. Hence, Tregs represent an ideal therapeutic tool to control detrimental immune reactions. Based on solid pre-clinical results, investigators started testing the safety and efficacy of Treg-based therapies in humans. Despite promising results, a number of issues remain to be solved. We will discuss the results obtained from clinical trials and the challenges and risks we are facing in the further development of Treg-based therapies.
Collapse
Affiliation(s)
- Silvia Gregori
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (TIGET) , Milan , Italy
| | - Laura Passerini
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, San Raffaele Telethon Institute for Gene Therapy (TIGET) , Milan , Italy
| | - Maria-Grazia Roncarolo
- Department of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine , Palo Alto, CA , USA
| |
Collapse
|
186
|
Scottà C, Fanelli G, Hoong SJ, Romano M, Lamperti EN, Sukthankar M, Guggino G, Fazekasova H, Ratnasothy K, Becker PD, Afzali B, Lechler RI, Lombardi G. Impact of immunosuppressive drugs on the therapeutic efficacy of ex vivo expanded human regulatory T cells. Haematologica 2015; 101:91-100. [PMID: 26471483 DOI: 10.3324/haematol.2015.128934] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 10/09/2015] [Indexed: 12/21/2022] Open
Abstract
Immunosuppressive drugs in clinical transplantation are necessary to inhibit the immune response to donor antigens. Although they are effective in controlling acute rejection, they do not prevent long-term transplant loss from chronic rejection. In addition, immunosuppressive drugs have adverse side effects, including increased rate of infections and malignancies. Adoptive cell therapy with human Tregs represents a promising strategy for the induction of transplantation tolerance. Phase I/II clinical trials in transplanted patients are already underway, involving the infusion of Tregs alongside concurrent immunosuppressive drugs. However, it remains to be determined whether the presence of immunosuppressive drugs negatively impacts Treg function and stability. We tested in vitro and in vivo the effects of tacrolimus, mycophenolate and methylprednisolone (major ISDs used in transplantation) on ex vivo expanded, rapamycin-treated human Tregs. The in vitro results showed that these drugs had no effect on phenotype, function and stability of Tregs, although tacrolimus affected the expression of chemokine receptors and IL-10 production. However, viability and proliferative capacity were reduced in a dose-dependent manner by all the three drugs. The in vivo experiments using a humanized mouse model confirmed the in vitro results. However, treatment of mice with only rapamycin maintained the viability, function and proliferative ability of adoptively transferred Tregs. Taken together, our results suggest that the key functions of ex vivo expanded Tregs are not affected by a concurrent immunosuppressive therapy. However, the choice of the drug combination and their timing and dosing should be considered as an essential component to induce and maintain tolerance by Treg.
Collapse
Affiliation(s)
- Cristiano Scottà
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giorgia Fanelli
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Sec Julie Hoong
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Marco Romano
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. & A. Seràgnoli", University of Bologna, Italy
| | - Estefania Nova Lamperti
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Mitalee Sukthankar
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giuliana Guggino
- Dipartimento di Biopatologia e Biotecnologie Mediche, University of Palermo, Italy
| | - Henrieta Fazekasova
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Kulachelvy Ratnasothy
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Pablo D Becker
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Behdad Afzali
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert I Lechler
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| | - Giovanna Lombardi
- Immunoregulation Laboratory, Division of Transplantation Immunology & Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, UK
| |
Collapse
|
187
|
Hall BM, Tran GT, Robinson CM, Hodgkinson SJ. Induction of antigen specific CD4+CD25+Foxp3+T regulatory cells from naïve natural thymic derived T regulatory cells. Int Immunopharmacol 2015; 28:875-86. [DOI: 10.1016/j.intimp.2015.03.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
|
188
|
Trzonkowski P, Bacchetta R, Battaglia M, Berglund D, Bohnenkamp HR, ten Brinke A, Bushell A, Cools N, Geissler EK, Gregori S, Marieke van Ham S, Hilkens C, Hutchinson JA, Lombardi G, Madrigal JA, Marek-Trzonkowska N, Martinez-Caceres EM, Roncarolo MG, Sanchez-Ramon S, Saudemont A, Sawitzki B. Hurdles in therapy with regulatory T cells. Sci Transl Med 2015; 7:304ps18. [PMID: 26355029 DOI: 10.1126/scitranslmed.aaa7721] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Improper activation of the immune system contributes to a variety of clinical conditions, including autoimmune and allergic diseases as well as solid organ and bone marrow transplantation. One approach to counteract this activation is through adoptive therapy with regulatory T cells (Tregs). Efforts to manufacture these cells have led to good maunfacturing practice-compliant protocols, and Treg products are entering early clinical trials. Here, we report the stance of the European Union Cooperation in Science and Technology Action BM1305, "Action to Focus and Accelerate Cell-based Tolerance-inducing Therapies-A FACTT," which identifies hurdles hindering Treg clinical applications in Europe and provides possible solutions.
Collapse
Affiliation(s)
- Piotr Trzonkowski
- Medical University of Gdansk, Department of Clinical Immunology and Transplantology, Debinki 7, 80-952 Gdansk, Poland. All authors equally contributed to this work.
| | - Rosa Bacchetta
- Department of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Standford, California, USA
| | - Manuela Battaglia
- Diabetes Research Institute (DRI), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - David Berglund
- Uppsala University, Department of Immunology, Genetics and Pathology; Section of Clinical Immunology, Rudbeck Laboratory, 751 85 Uppsala, Sweden
| | | | - Anja ten Brinke
- Department of Immunopathology, Sanquin Blood Supply, Division Research, Plesmanlaan 125, 1066 CX Amsterdam, Netherland and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Andrew Bushell
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, B-2650 Edegem, Belgium
| | - Edward K Geissler
- Division of Experimental Surgery, Department of Surgery, University Hospital Regensburg, Regensburg, Bavaria, 93053, Germany
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Blood Supply, Division Research, Plesmanlaan 125, 1066 CX Amsterdam, Netherland and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | | | - James A Hutchinson
- Division of Experimental Surgery, Department of Surgery, University Hospital Regensburg, Regensburg, 93053, Bavaria, Germany
| | - Giovanna Lombardi
- Medical Research Council (MRC) Centre in Transplantation, Kings College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - J Alejandro Madrigal
- Anthony Nolan Research Institute, University College London (UCL) Royal Free Hospital Campus, 77c Fleet Road, London NW3 2QG, UK
| | | | - Eva M Martinez-Caceres
- Immunology Division, Germans Trias i Pujol University Hospital. Campus Can Ruti. Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma Barcelona 08916, Badalona, Barcelona, Spain
| | - Maria Grazia Roncarolo
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy. Department of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Stanford, California, USA
| | - Silvia Sanchez-Ramon
- Departamento de Inmunología Clínica, Hospital Clínico San Carlos, Calle Profesor Martín Lagos S/N, E- 28040 Madrid, Spain
| | - Aurore Saudemont
- Anthony Nolan Research Institute, University College London (UCL) Royal Free Hospital Campus, 77c Fleet Road, London NW3 2QG, UK
| | - Birgit Sawitzki
- AG Transplantationstoleranz, Charite Universitätsmedizin, Institut für Med. Imunologie, Augustenburgerplatz 1, 13353 Berlin, Germany
| |
Collapse
|
189
|
Safinia N, Scotta C, Vaikunthanathan T, Lechler RI, Lombardi G. Regulatory T Cells: Serious Contenders in the Promise for Immunological Tolerance in Transplantation. Front Immunol 2015; 6:438. [PMID: 26379673 PMCID: PMC4553385 DOI: 10.3389/fimmu.2015.00438] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/12/2015] [Indexed: 01/12/2023] Open
Abstract
Regulatory T cells (Tregs) play an important role in immunoregulation and have been shown in animal models to promote transplantation tolerance and curb autoimmunity following their adoptive transfer. The safety and potential therapeutic efficacy of these cells has already been reported in Phase I trials of bone-marrow transplantation and type I diabetes, the success of which has motivated the broadened application of these cells in solid-organ transplantation. Despite major advances in the clinical translation of these cells, there are still key questions to be addressed to ensure that Tregs attest their reputation as ideal candidates for tolerance induction. In this review, we will discuss the unique traits of Tregs that have attracted such fame in the arena of tolerance induction. We will outline the protocols used for their ex vivo expansion and discuss the future directions of Treg cell therapy. In this regard, we will review the concept of Treg heterogeneity, the desire to isolate and expand a functionally superior Treg population and report on the effect of differing culture conditions. The relevance of Treg migratory capacity will also be discussed together with methods of in vivo visualization of the infused cells. Moreover, we will highlight key advances in the identification and expansion of antigen-specific Tregs and discuss their significance for cell therapy application. We will also summarize the clinical parameters that are of importance, alongside cell manufacture, from the choice of immunosuppression regimens to the number of injections in order to direct the success of future efficacy trials of Treg cell therapy. Years of research in the field of tolerance have seen an accumulation of knowledge and expertise in the field of Treg biology. This perpetual progression has been the driving force behind the many successes to date and has put us now within touching distance of our ultimate success, immunological tolerance.
Collapse
Affiliation(s)
- Niloufar Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Cristiano Scotta
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Trishan Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Robert I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London , London , UK
| |
Collapse
|
190
|
Abstract
PURPOSE OF REVIEW CD4Foxp3 regulatory T cells (Tregs) are crucial in controlling immunity and self-tolerance. Consequently, in transplantation, Tregs play a central role in inhibiting acute rejection and promoting allograft tolerance. A more complete understanding of Treg biology may lead to novel therapeutic approaches to enhance Treg numbers and function. RECENT FINDINGS The maintenance of self-tolerance in nonlymphoid tissues requires the differentiation of Tregs in secondary lymphoid organs from naïve-like central Tregs into effector Tregs. Antigen and environmental cues guide this Treg differentiation, which parallels the types of adaptive immune responses taking place, allowing them to enter and function within specific nonlymphoid tissues. In addition to controlling inflammation, tissue-infiltrating Tregs unexpectedly regulate nonimmune processes, including metabolic homeostasis and tissue repair. Finally, Tregs can be directly and specifically targeted in vivo to augment their numbers or enhance their function in both secondary lymphoid organs and nonlymphoid tissues. SUMMARY Tregs exhibit a previously unrecognized breadth of function, which includes tissue-specific specialization and the regulation of both immune and nonimmune processes. This is of particular importance in transplantation since allo-reactive memory T cells can act directly within the allograft. Thus, therapeutic approaches may need to promote Treg function in transplanted tissue, as well as in secondary lymphoid organs. Such therapy would not only prevent inflammation and acute rejection, but may also promote nonimmune processes within the allograft such as tissue homeostasis and repair.
Collapse
Affiliation(s)
- David M. Rothstein
- Departments of Medicine and Immunology, University of Pittsburgh Medical School, The Thomas E. Starzl Transplantation Institute, Pittsburgh PA
- Department of Surgery, University of Pittsburgh Medical School, The Thomas E. Starzl Transplantation Institute, Pittsburgh PA
| | - Geoffrey Camirand
- Department of Surgery, University of Pittsburgh Medical School, The Thomas E. Starzl Transplantation Institute, Pittsburgh PA
| |
Collapse
|
191
|
Mason GM, Lowe K, Melchiotti R, Ellis R, de Rinaldis E, Peakman M, Heck S, Lombardi G, Tree TIM. Phenotypic Complexity of the Human Regulatory T Cell Compartment Revealed by Mass Cytometry. THE JOURNAL OF IMMUNOLOGY 2015. [DOI: 10.4049/jimmunol.1500703] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
192
|
Niemann N, Sawitzki B. Treg Therapy in Transplantation: How and When Will We Do It? CURRENT TRANSPLANTATION REPORTS 2015. [DOI: 10.1007/s40472-015-0066-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
193
|
You S. Differential sensitivity of regulatory and effector T cells to cell death: a prerequisite for transplant tolerance. Front Immunol 2015; 6:242. [PMID: 26042125 PMCID: PMC4437185 DOI: 10.3389/fimmu.2015.00242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/06/2015] [Indexed: 12/13/2022] Open
Abstract
Despite significant progress achieved in transplantation, immunosuppressive therapies currently used to prevent graft rejection are still endowed with severe side effects impairing their efficiency over the long term. Thus, the development of graft-specific, non-toxic innovative therapeutic strategies has become a major challenge, the goal being to selectively target alloreactive effector T cells while sparing CD4+Foxp3+ regulatory T cells (Tregs) to promote operational tolerance. Various approaches, notably the one based on monoclonal antibodies or fusion proteins directed against the TCR/CD3 complex, TCR coreceptors, or costimulatory molecules, have been proposed to reduce the alloreactive T cell pool, which is an essential prerequisite to create a therapeutic window allowing Tregs to induce and maintain allograft tolerance. In this mini review, we focus on the differential sensitivity of Tregs and effector T cells to the depleting and inhibitory effect of these immunotherapies, with a particular emphasis on CD3-specific antibodies that beyond their immunosuppressive effect, also express potent tolerogenic capacities.
Collapse
Affiliation(s)
- Sylvaine You
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; INSERM U1151, Institut Necker-Enfants Malades , Paris , France ; CNRS UMR 8253, Institut Necker-Enfants Malades , Paris , France
| |
Collapse
|
194
|
Bluestone JA, Bour-Jordan H, Cheng M, Anderson M. T cells in the control of organ-specific autoimmunity. J Clin Invest 2015; 125:2250-60. [PMID: 25985270 DOI: 10.1172/jci78089] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Immune tolerance is critical to the avoidance of unwarranted immune responses against self antigens. Multiple, non-redundant checkpoints are in place to prevent such potentially deleterious autoimmune responses while preserving immunity integral to the fight against foreign pathogens. Nevertheless, a large and growing segment of the population is developing autoimmune diseases. Deciphering cellular and molecular pathways of immune tolerance is an important goal, with the expectation that understanding these pathways will lead to new clinical advances in the treatment of these devastating diseases. The vast majority of autoimmune diseases develop as a consequence of complex mechanisms that depend on genetic, epigenetic, molecular, cellular, and environmental elements and result in alterations in many different checkpoints of tolerance and ultimately in the breakdown of immune tolerance. The manifestations of this breakdown are harmful inflammatory responses in peripheral tissues driven by innate immunity and self antigen-specific pathogenic T and B cells. T cells play a central role in the regulation and initiation of these responses. In this Review we summarize our current understanding of the mechanisms involved in these fundamental checkpoints, the pathways that are defective in autoimmune diseases, and the therapeutic strategies being developed with the goal of restoring immune tolerance.
Collapse
|
195
|
Zhang H, Guo H, Lu L, Zahorchak AF, Wiseman RW, Raimondi G, Cooper DKC, Ezzelarab MB, Thomson AW. Sequential monitoring and stability of ex vivo-expanded autologous and nonautologous regulatory T cells following infusion in nonhuman primates. Am J Transplant 2015; 15:1253-66. [PMID: 25783759 PMCID: PMC4773915 DOI: 10.1111/ajt.13113] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 10/24/2014] [Accepted: 11/17/2014] [Indexed: 01/25/2023]
Abstract
Ex vivo-expanded cynomolgus monkey CD4(+)CD25(+)CD127(-) regulatory T cells (Treg) maintained Foxp3 demethylation status at the Treg-specific demethylation region, and potently suppressed T cell proliferation through three rounds of expansion. When carboxyfluorescein succinimidyl ester- or violet proliferation dye 450-labeled autologous (auto) and nonautologous (non-auto)-expanded Treg were infused into monkeys, the number of labeled auto-Treg in peripheral blood declined rapidly during the first week, but persisted at low levels in both normal and anti-thymocyte globulin plus rapamycin-treated (immunosuppressed; IS) animals for at least 3 weeks. By contrast, MHC-mismatched non-auto-Treg could not be detected in normal monkey blood or in blood of two out of the three IS monkeys by day 6 postinfusion. They were also more difficult to detect than auto-Treg in peripheral lymphoid tissue. Both auto- and non-auto-Treg maintained Ki67 expression early after infusion. Sequential monitoring revealed that adoptively transferred auto-Treg maintained similarly high levels of Foxp3 and CD25 and low CD127 compared with endogenous Treg, although Foxp3 staining diminished over time in these nontransplanted recipients. Thus, infused ex vivo-expanded auto-Treg persist longer than MHC-mismatched non-auto-Treg in blood of nonhuman primates and can be detected in secondary lymphoid tissue. Host lymphodepletion and rapamycin administration did not consistently prolong the persistence of non-auto-Treg in these sites.
Collapse
Affiliation(s)
- H. Zhang
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - H. Guo
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Lu
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. F. Zahorchak
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - R. W. Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI
| | - G. Raimondi
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D. K. C. Cooper
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. B. Ezzelarab
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. W. Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Angus W. Thomson,
| |
Collapse
|
196
|
Litjens NHR, Boer K, Zuijderwijk JM, Klepper M, Peeters AMA, Prens EP, Verschoor W, Kraaijeveld R, Ozgur Z, van den Hout-van Vroonhoven MC, van IJcken WFJ, Baan CC, Betjes MGH. Allogeneic Mature Human Dendritic Cells Generate Superior Alloreactive Regulatory T Cells in the Presence of IL-15. THE JOURNAL OF IMMUNOLOGY 2015; 194:5282-93. [PMID: 25917092 DOI: 10.4049/jimmunol.1402827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/23/2015] [Indexed: 11/19/2022]
Abstract
Expansion of Ag-specific naturally occurring regulatory T cells (nTregs) is required to obtain sufficient numbers of cells for cellular immunotherapy. In this study, different allogeneic stimuli were studied for their capacity to generate functional alloantigen-specific nTregs. A highly enriched nTreg fraction (CD4(+)CD25(bright)CD127(-) T cells) was alloantigen-specific expanded using HLA-mismatched immature, mature monocyte-derived dendritic cells (moDCs), or PBMCs. The allogeneic mature moDC-expanded nTregs were fully characterized by analysis of the demethylation status within the Treg-specific demethylation region of the FOXP3 gene and the expression of both protein and mRNA of FOXP3, HELIOS, CTLA4, and cytokines. In addition, the Ag-specific suppressive capacity of these expanded nTregs was tested. Allogeneic mature moDCs and skin-derived DCs were superior in inducing nTreg expansion compared with immature moDCs or PBMCs in an HLA-DR- and CD80/CD86-dependent way. Remarkably, the presence of exogenous IL-15 without IL-2 could facilitate optimal mature moDC-induced nTreg expansion. Allogeneic mature moDC-expanded nTregs were at low ratios (<1:320), potent suppressors of alloantigen-induced proliferation without significant suppression of completely HLA-mismatched, Ag-induced proliferation. Mature moDC-expanded nTregs were highly demethylated at the Treg-specific demethylation region within the FOXP3 gene and highly expressed of FOXP3, HELIOS, and CTLA4. A minority of the expanded nTregs produced IL-10, IL-2, IFN-γ, and TNF-α, but few IL-17-producing nTregs were found. Next-generation sequencing of mRNA of moDC-expanded nTregs revealed a strong induction of Treg-associated mRNAs. Human allogeneic mature moDCs are highly efficient stimulator cells, in the presence of exogenous IL-15, for expansion of stable alloantigen-specific nTregs with superior suppressive function.
Collapse
Affiliation(s)
- Nicolle H R Litjens
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands;
| | - Karin Boer
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Joke M Zuijderwijk
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Mariska Klepper
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Annemiek M A Peeters
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Errol P Prens
- Department of Dermatology, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands; Department of Rheumatology, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands; and
| | - Wenda Verschoor
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Rens Kraaijeveld
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Zeliha Ozgur
- Erasmus Medical Center, Erasmus Center for Biomics, 3000 CA Rotterdam, the Netherlands
| | | | | | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3000 CA Rotterdam, the Netherlands
| |
Collapse
|
197
|
Tang Q, Kang SM. Interpretation of transplant biopsies and immune responses following Treg cell therapy. Curr Opin Organ Transplant 2015; 19:616-20. [PMID: 25313874 DOI: 10.1097/mot.0000000000000132] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Regulatory T cells (Treg) are now well established as vital participants in maintaining self-tolerance and preventing autoimmunity. Tregs have already been shown to be effective in preventing graft-versus-host disease in clinical bone marrow transplantation, and numerous animal studies have suggested a therapeutic role for Treg in solid organ transplantation. Recent advances in Treg isolation and expansion have the field poised to perform trials of therapeutic Treg infusion in solid organ transplantation worldwide. An important component of these trials will be the detection of infused cells and the assessment of Treg activity after infusion. RECENT FINDINGS Several animal studies have demonstrated that infused Treg migrate to transplanted tissue in the early period after transplantation. This finding has important implications for the interpretation of biopsy results in human trials. Recent refinements in Treg identification, quantification, and functional assays will be discussed in the context of immune monitoring. SUMMARY Understanding the migration/localization and persistence of infused Treg into transplanted tissues as well as how they impact the peripheral immune response will be critical to the interpretation of early Treg trials.
Collapse
Affiliation(s)
- Qizhi Tang
- Department of Surgery, University of California, San Francisco, California, USA
| | | |
Collapse
|
198
|
Abstract
PURPOSE OF REVIEW Cellular therapies show early proof of safety and efficacy in a, so far, limited number of clinical trials. Technical and regulatory difficulties in translating innovative preclinical cell therapy approaches into clinical protocols have been and still are a major roadblock for a more rapid progress. This is particularly true for broad clinical applications of cellular therapies outside specialized clinical centers and for the initiation of multicenter clinical trials. The increased awareness of such deficits in availability of cGMP-compliant technologies and integration of multistep procedures into one clinical manufacturing process has initiated efforts from both basic researchers and biotech companies to overcome these problems. These developments will be exemplified with a focus on T-cell therapies and, in particular, the use of regulatory T cells for bone marrow and organ transplantation or autoimmunity. RECENT FINDINGS Recent developments in the field of clinical cell sorting, identification of therapeutically relevant T-cell subsets, in-vitro cell culture technologies and automation have the potential to provide solutions for long-standing problems in the field of clinical cell processing. SUMMARY Recent technological developments and further attempts aiming at simplification, integration of multistep procedures and automation of clinical cell processing are key for the development of successful cellular therapies and their broad implementation in the clinics.
Collapse
|
199
|
The potential role for regulatory T-cell therapy in vascularized composite allograft transplantation. Curr Opin Organ Transplant 2015; 19:558-65. [PMID: 25333829 DOI: 10.1097/mot.0000000000000139] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Vascularized composite allograft (VCA) transplantation restores defects to a degree not possible by conventional techniques. However, it is limited by the need for long-term immunosuppression and high rates of acute rejection directed against skin. There is therefore a need for a therapy that may shift the risk-benefit ratio in favour of VCA transplantation. Regulatory T cells (Tregs) are a subset of T cells with potent immunoregulatory properties and the potential to promote immunosuppression-free allograft survival. In this review, we consider the evidence for Treg therapy in VCA transplantation. RECENT FINDINGS CD4 Tregs are the best-studied immunoregulatory cell type, and a large amount of experimental and clinical data is emerging to endorse their use in VCA transplantation. Data from animal and humanized models are particularly encouraging and demonstrate the potent efficacy of Treg at preventing skin allograft rejection. Moreover, central tolerance induction techniques in VCA transplantation models are demonstrating a dependence on Tregs for graft survival. SUMMARY An improvement in outcomes after VCA transplantation has the potential to revolutionize the field. Several effective therapeutic strategies have demonstrated great promise experimentally, and there is now a need to assess their safety and efficacy in a clinical setting.
Collapse
|
200
|
Bluestone JA, Trotta E, Xu D. The therapeutic potential of regulatory T cells for the treatment of autoimmune disease. Expert Opin Ther Targets 2015; 19:1091-103. [PMID: 25881491 DOI: 10.1517/14728222.2015.1037282] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Immune tolerance remains the holy grail of therapeutic immunology in the fields of organ and tissue transplant rejection, autoimmune diseases, and allergy and asthma. We have learned that FoxP3(+)CD4(+) regulatory T cells play a vital role in both the induction and maintenance of self-tolerance. AREAS COVERED In this opinion piece, we highlight regulatory T cells (Treg) cell biology and novel immune treatments to take advantage of these cells as potent therapeutics. We discuss the potential to utilize Treg and Treg-friendly therapies to replace current general immunosuppressives and induce tolerance as a path towards a drug-free existence without associated toxicities. EXPERT OPINION Finally, we opine on the fact that biomedicine sits on the cusp of a new revolution: the use of human cells as versatile therapeutic engines. We highlight the challenges and opportunities associated with the development of a foundational cellular engineering science that provides a systematic framework for safely and predictably regulating cellular behaviors. Although Treg therapy has become a legitimate clinical treatment, development of the therapy will require a better understanding of the underlying Treg biology, manufacturing advances to promote cost effectiveness and combinations with other drugs to alter the pathogenicity/regulatory balance.
Collapse
Affiliation(s)
- Jeffrey A Bluestone
- University of California San Francisco, Diabetes Center , San Francisco, CA 94143 , USA +1 415 476 4451 ; jeff,
| | | | | |
Collapse
|