201
|
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: 367] [Impact Index Per Article: 40.8] [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
|
202
|
Scalea JR, Tomita Y, Lindholm CR, Burlingham W. Transplantation Tolerance Induction: Cell Therapies and Their Mechanisms. Front Immunol 2016; 7:87. [PMID: 27014267 PMCID: PMC4779899 DOI: 10.3389/fimmu.2016.00087] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/22/2016] [Indexed: 12/12/2022] Open
Abstract
Cell-based therapies have been studied extensively in the context of transplantation tolerance induction. The most successful protocols have relied on transfusion of bone marrow prior to the transplantation of a renal allograft. However, it is not clear that stem cells found in bone marrow are required in order to render a transplant candidate immunologically tolerant. Accordingly, mesenchymal stem cells, regulatory myeloid cells, T regulatory cells, and other cell types are being tested as possible routes to tolerance induction, in the absence of donor-derived stem cells. Early data with each of these cell types have been encouraging. However, the induction regimen capable of achieving consistent tolerance, while avoiding unwanted sided effects, and which is scalable to the human patient, has yet to be identified. Here, we present the status of investigations of various tolerogenic cell types and the mechanistic rationale for their use in tolerance induction protocols.
Collapse
Affiliation(s)
- Joseph R Scalea
- Department of Surgery, Division of Transplantation, University of Wiconsin , Madison, WI , USA
| | - Yusuke Tomita
- Department of Surgery, Division of Transplantation, University of Wiconsin , Madison, WI , USA
| | | | - William Burlingham
- Department of Surgery, Division of Transplantation, University of Wiconsin , Madison, WI , USA
| |
Collapse
|
203
|
Garnier A, Hamieh M, Drouet A, Leprince J, Vivien D, Frébourg T, Le Mauff B, Latouche JB, Toutirais O. Artificial antigen-presenting cells expressing HLA class II molecules as an effective tool for amplifying human specific memory CD4(+) T cells. Immunol Cell Biol 2016; 94:662-72. [PMID: 26924643 DOI: 10.1038/icb.2016.25] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 02/07/2023]
Abstract
Owing to their multiple immune functions, CD4(+) T cells are of major interest for immunotherapy in chronic viral infections and cancer, as well as for severe autoimmune diseases and transplantation. Therefore, standardized methods allowing rapid generation of a large number of CD4(+) T cells for adoptive immunotherapy are still awaited. We constructed stable artificial antigen-presenting cells (AAPCs) derived from mouse fibroblasts. They were genetically modified to express human leukocyte antigen (HLA)-DR molecules and the human accessory molecules B7.1, Intercellular adhesion molecule-1 (ICAM-1) and lymphocyte function-associated antigen-3 (LFA-3). AAPCs expressing HLA-DR1, HLA-DR15 or HLA-DR51 molecules and loaded with peptides derived from influenza hemagglutinin (HA), myelin basic protein (MBP) or factor VIII, respectively, activated specific CD4(+) T-cell clones more effectively than Epstein-Barr virus (EBV)-transformed B cells. We also showed that AAPCs were able to take up and process whole Ag proteins, and present epitopes to specific T cells. In primary cultures, AAPCs loaded with HA peptide allowed generation of specific Th1 lymphocytes from healthy donors as demonstrated by tetramer and intracellular cytokine staining. Although AAPCs were less effective than autologous peripheral blood mononuclear cells (PBMCs) to stimulate CD4(+) T cells in primary culture, AAPCs were more potent to reactivate and expand memory Th1 cells in a strictly Ag-dependent manner. As the availability of autologous APCs is limited, the AAPC system represents a stable and reliable tool to achieve clinically relevant numbers of CD4(+) T cells for adoptive immunotherapy. For fundamental research in immunology, AAPCs are also useful to decipher mechanisms involved in the development of human CD4 T-cell responses.
Collapse
Affiliation(s)
- Anthony Garnier
- Inserm U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Caen, France.,Université de Caen Basse-Normandie, UFR Médecine, Caen, France
| | - Mohamad Hamieh
- Inserm U1079, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Rouen, France
| | - Aurélie Drouet
- Inserm U1079, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Rouen, France
| | - Jérôme Leprince
- Inserm U982, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Rouen, France
| | - Denis Vivien
- Inserm U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Caen, France.,Université de Caen Basse-Normandie, UFR Médecine, Caen, France
| | - Thierry Frébourg
- Inserm U1079, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - Brigitte Le Mauff
- Inserm U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Caen, France.,Université de Caen Basse-Normandie, UFR Médecine, Caen, France.,CHU Caen, Department of Immunology and Immunopathology, Caen, France.,Etablissement Français du Sang, Normandie, Caen, France
| | - Jean-Baptiste Latouche
- Inserm U1079, Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - Olivier Toutirais
- Inserm U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Caen, France.,Université de Caen Basse-Normandie, UFR Médecine, Caen, France.,CHU Caen, Department of Immunology and Immunopathology, Caen, France.,Etablissement Français du Sang, Normandie, Caen, France
| |
Collapse
|
204
|
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
|
205
|
Sicard A, Koenig A, Graff-Dubois S, Dussurgey S, Rouers A, Dubois V, Blanc P, Chartoire D, Errazuriz-Cerda E, Paidassi H, Taillardet M, Morelon E, Moris A, Defrance T, Thaunat O. B Cells Loaded with Synthetic Particulate Antigens: A Versatile Platform To Generate Antigen-Specific Helper T Cells for Cell Therapy. NANO LETTERS 2016; 16:297-308. [PMID: 26650819 DOI: 10.1021/acs.nanolett.5b03801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Adoptive cell therapy represents a promising approach for several chronic diseases. This study describes an innovative strategy for biofunctionalization of nanoparticles, allowing the generation of synthetic particulate antigens (SPAg). SPAg activate polyclonal B cells and vectorize noncognate proteins into their endosomes, generating highly efficient stimulators for ex vivo expansion of antigen-specific CD4+ T cells. This method also allows harnessing the ability of B cells to polarize CD4+ T cells into effectors or regulators.
Collapse
Affiliation(s)
- Antoine Sicard
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
- Edouard Herriot Hospital , Transplantation, Nephrology and Clinical Immunology Department, 69003 Lyon, France
| | - Alice Koenig
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
- Edouard Herriot Hospital , Transplantation, Nephrology and Clinical Immunology Department, 69003 Lyon, France
| | - Stéphanie Graff-Dubois
- Sorbonne University , UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Center for Immunology and Microbial Infections - CIMI-Paris, F-75013, Paris, France
| | - Sébastien Dussurgey
- SFR Biosciences, UMS344/US8, Inserm, CNRS, Claude Bernard Lyon-1 University, Ecole Normale Supérieure , 69007 Lyon, France
| | - Angéline Rouers
- Sorbonne University , UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Center for Immunology and Microbial Infections - CIMI-Paris, F-75013, Paris, France
| | - Valérie Dubois
- French National Blood Service (EFS) , 69007 Lyon, France
| | - Pascal Blanc
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
| | - Dimitri Chartoire
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
| | | | - Helena Paidassi
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
| | - Morgan Taillardet
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
| | - Emmanuel Morelon
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
- Edouard Herriot Hospital , Transplantation, Nephrology and Clinical Immunology Department, 69003 Lyon, France
| | - Arnaud Moris
- Sorbonne University , UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Center for Immunology and Microbial Infections - CIMI-Paris, F-75013, Paris, France
| | - Thierry Defrance
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
| | - Olivier Thaunat
- International Center for Infectiology Research (CIRI); French National Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard Lyon 1 University; Ecole Normale Supérieure de Lyon, CNRS, UMR 5308 , 69007 Lyon, France
- Edouard Herriot Hospital , Transplantation, Nephrology and Clinical Immunology Department, 69003 Lyon, France
| |
Collapse
|
206
|
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
|
207
|
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
|
208
|
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
|
209
|
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: 51] [Impact Index Per Article: 5.1] [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
|
210
|
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]
|
211
|
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
|
212
|
Thymus-Derived Regulatory T Cells Infiltrate the Cardiac Allograft Before Rejection. Transplantation 2015; 99:1839-46. [DOI: 10.1097/tp.0000000000000730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
213
|
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: 96] [Impact Index Per Article: 9.6] [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
|
214
|
Yang EY, Kronenfeld JP, Gattás-Asfura KM, Bayer AL, Stabler CL. Engineering an "infectious" T(reg) biomimetic through chemoselective tethering of TGF-β1 to PEG brush surfaces. Biomaterials 2015. [PMID: 26197412 DOI: 10.1016/j.biomaterials.2015.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Modulation of immunological responses to allografts following transplantation is of pivotal importance to improving graft outcome and duration. Of the many approaches, harnessing the dominant tolerance induced by regulatory T cells (Treg) holds tremendous promise. Recent studies have highlighted the unique potency of cell surface-bound TGF-β1 on Treg for promoting infectious tolerance, i.e. to confer suppressive capacity from one cell to another. To mimic this characteristic, TGF-β1 was chemoselectively tethered to inert and viable polymer grafting platforms using Staudinger ligation. We report the synthesis and functional characterization of these engineered TGF-β1 surfaces. Inert beads tethered with TGF-β1 were capable of efficiently converting naïve CD4(+) CD62L(hi) T cells to functional Treg. Concordantly, translation of conjugation scheme from inert surfaces to viable cells also led to efficient generation of functional Treg. Further, the capacity of these platforms to generate antigen-specific Treg was demonstrated. These findings illustrate the unique faculty of tethered TGF-β1 biomaterial platforms to function as an "infectious" Treg and provide a compelling approach for generating tolerogenic microenvironments for allograft transplantation.
Collapse
Affiliation(s)
- E Y Yang
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
| | - J P Kronenfeld
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Medicine, University of Miami, Miami, FL, USA
| | | | - A L Bayer
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Microbiology and Immunology, University of Miami, Miami, FL, USA
| | - C L Stabler
- Diabetes Research Institute, University of Miami, Miami, FL, USA; Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA; Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Surgery, University of Miami, Miami, FL, USA.
| |
Collapse
|
215
|
Verhagen J, Wegner A, Wraith DC. Extra-thymically induced T regulatory cell subsets: the optimal target for antigen-specific immunotherapy. Immunology 2015; 145:171-81. [PMID: 25716063 DOI: 10.1111/imm.12458] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 12/13/2022] Open
Abstract
Antigen-specific immunotherapy aims to selectively restore tolerance to innocuous antigens in cases of autoimmune or allergic disease, without the need for general immune suppression. Although the principle of antigen-specific immunotherapy was discovered more than a century ago, its clinical application to date is limited, particularly in the control of autoimmunity. This has resulted mainly from a lack of in-depth understanding of the underlying mechanism. More recently, the differentiation of extra-thymically induced T regulatory (Treg) cell subsets has been shown to be instrumental in peripheral tolerance induction. Two main types of inducible Treg cells, interleukin-10-secreting or Foxp3(+) , have now been described, each with distinct characteristics and methods of therapeutic induction. It is crucial, therefore, to identify the suitability of either subset in the control of specific immune disorders. This review explores their natural function, the known mechanisms of therapeutic differentiation of either subset as well as their in vivo functionality and discusses new developments that may aid their use in antigen-specific immunotherapy, with a focus on autoimmune disease.
Collapse
Affiliation(s)
- Johan Verhagen
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | | | |
Collapse
|
216
|
Koboziev I, Jones-Hall Y, Valentine JF, Webb CR, Furr KL, Grisham MB. Use of Humanized Mice to Study the Pathogenesis of Autoimmune and Inflammatory Diseases. Inflamm Bowel Dis 2015; 21:1652-73. [PMID: 26035036 PMCID: PMC4466023 DOI: 10.1097/mib.0000000000000446] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Animal models of disease have been used extensively by the research community for the past several decades to better understand the pathogenesis of different diseases and assess the efficacy and toxicity of different therapeutic agents. Retrospective analyses of numerous preclinical intervention studies using mouse models of acute and chronic inflammatory diseases reveal a generalized failure to translate promising interventions or therapeutics into clinically effective treatments in patients. Although several possible reasons have been suggested to account for this generalized failure to translate therapeutic efficacy from the laboratory bench to the patient's bedside, it is becoming increasingly apparent that the mouse immune system is substantially different from the human. Indeed, it is well known that >80 major differences exist between mouse and human immunology; all of which contribute to significant differences in immune system development, activation, and responses to challenges in innate and adaptive immunity. This inconvenient reality has prompted investigators to attempt to humanize the mouse immune system to address important human-specific questions that are impossible to study in patients. The successful long-term engraftment of human hematolymphoid cells in mice would provide investigators with a relatively inexpensive small animal model to study clinically relevant mechanisms and facilitate the evaluation of human-specific therapies in vivo. The discovery that targeted mutation of the IL-2 receptor common gamma chain in lymphopenic mice allows for the long-term engraftment of functional human immune cells has advanced greatly our ability to humanize the mouse immune system. The objective of this review is to present a brief overview of the recent advances that have been made in the development and use of humanized mice with special emphasis on autoimmune and chronic inflammatory diseases. In addition, we discuss the use of these unique mouse models to define the human-specific immunopathological mechanisms responsible for the induction and perpetuation of chronic gut inflammation.
Collapse
Affiliation(s)
- Iurii Koboziev
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Yava Jones-Hall
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN 47907-2027
| | - John F. Valentine
- Department of Internal Medicine, Gastroenterology, Hepatology and Nutrition, University of Utah, Salt Lake City, UT 84132-2410
| | - Cynthia Reinoso Webb
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| |
Collapse
|
217
|
Sanmamed MF, Rodriguez I, Schalper KA, Oñate C, Azpilikueta A, Rodriguez-Ruiz ME, Morales-Kastresana A, Labiano S, Pérez-Gracia JL, Martín-Algarra S, Alfaro C, Mazzolini G, Sarno F, Hidalgo M, Korman AJ, Jure-Kunkel M, Melero I. Nivolumab and Urelumab Enhance Antitumor Activity of Human T Lymphocytes Engrafted in Rag2-/-IL2Rγnull Immunodeficient Mice. Cancer Res 2015; 75:3466-78. [PMID: 26113085 DOI: 10.1158/0008-5472.can-14-3510] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 05/31/2015] [Indexed: 11/16/2022]
Abstract
A current pressing need in cancer immunology is the development of preclinical model systems that are immunocompetent for the study of human tumors. Here, we report the development of a humanized murine model that can be used to analyze the pharmacodynamics and antitumor properties of immunostimulatory monoclonal antibodies (mAb) in settings where the receptors targeted by the mAbs are expressed. Human lymphocytes transferred into immunodeficient mice underwent activation and redistribution to murine organs, where they exhibited cell-surface expression of hCD137 and hPD-1. Systemic lymphocyte infiltrations resulted in a lethal CD4(+) T cell-mediated disease (xenograft-versus-host disease), which was aggravated when murine subjects were administered clinical-grade anti-hCD137 (urelumab) and anti-hPD-1 (nivolumab). In mice engrafted with human colorectal HT-29 carcinoma cells and allogeneic human peripheral blood mononuclear cells (PBMC), or with a patient-derived gastric carcinoma and PBMCs from the same patient, we found that coadministration of urelumab and nivolumab was sufficient to significantly slow tumor growth. Correlated with this result were increased numbers of activated human T lymphocytes producing IFNγ and decreased numbers of human regulatory T lymphocytes in the tumor xenografts, possibly explaining the efficacy of the therapeutic regimen. Our results offer a proof of concept for the use of humanized mouse models for surrogate efficacy and histology investigations of immune checkpoint drugs and their combinations.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain. Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Inmaculada Rodriguez
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Carmen Oñate
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Arantza Azpilikueta
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain. Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | - Sara Labiano
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | | | - Carlos Alfaro
- Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Department of Medicine, Universidad Austral, Pilar, Argentina
| | - Francesca Sarno
- Centro Integral Oncológico Clara Campal (CIOCC), Madrid, Spain
| | - Manuel Hidalgo
- Centro Integral Oncológico Clara Campal (CIOCC), Madrid, Spain. Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alan J Korman
- Biologics Discovery California, Bristol-Myers Squibb, Redwood City, California
| | | | - Ignacio Melero
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain. Centro de investigación médica aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
218
|
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]
|
219
|
Morath C, Schmitt A, Zeier M, Schmitt M, Sandra-Petrescu F, Opelz G, Terness P, Schaier M, Kleist C. Cell therapy for immunosuppression after kidney transplantation. Langenbecks Arch Surg 2015; 400:541-50. [PMID: 26077202 DOI: 10.1007/s00423-015-1313-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/03/2015] [Indexed: 01/03/2023]
Abstract
PURPOSE To give an overview over cell therapeutic approaches to immunosuppression in clinical kidney transplantation. A focus is on myeloid suppressor cell therapy by mitomycin C-induced cells (MICs). METHODS Literature review with an emphasis on already existing therapies. RESULTS Several cell therapeutic approaches to immunosuppression and donor-specific unresponsiveness are now being tested in early phase I and phase II trials in clinical kidney transplantation. Cell products such as regulatory T cells or regulatory macrophages, or other myeloid suppressor cell therapies, may either consist of donor-specific, third-party, or autologous cell preparations. Major problems are the identification of the suppressive cell populations and their expansion to have sufficient amount of cells to achieve donor unresponsiveness (e.g., with regulatory T cells). We show a simple and safe way to establish donor unresponsiveness in living-donor kidney transplantation by MIC therapy. A phase I clinical trial is now under way to test the safety and efficacy of this cell therapeutic approach. CONCLUSIONS Cell therapeutic approaches to immunosuppression after kidney transplantation may revolutionize clinical transplantation in the future.
Collapse
Affiliation(s)
- Christian Morath
- Division of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
220
|
van der Net JB, Bushell A, Wood KJ, Harden PN. Regulatory T cells: first steps of clinical application in solid organ transplantation. Transpl Int 2015; 29:3-11. [DOI: 10.1111/tri.12608] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/26/2015] [Accepted: 05/13/2015] [Indexed: 01/03/2023]
Affiliation(s)
- Jeroen B. van der Net
- Transplantation Research Immunology Group; Nuffield Department of Surgical Sciences; University of Oxford; Oxford UK
- Oxford Transplant Centre; Oxford University Hospitals NHS Trust; Oxford UK
| | - Andrew Bushell
- Transplantation Research Immunology Group; Nuffield Department of Surgical Sciences; University of Oxford; Oxford UK
| | - Kathryn J. Wood
- Transplantation Research Immunology Group; Nuffield Department of Surgical Sciences; University of Oxford; Oxford UK
| | - Paul N. Harden
- Oxford Transplant Centre; Oxford University Hospitals NHS Trust; Oxford UK
| |
Collapse
|
221
|
Montano-Loza AJ, Czaja AJ. Cell mediators of autoimmune hepatitis and their therapeutic implications. Dig Dis Sci 2015; 60:1528-42. [PMID: 25487192 DOI: 10.1007/s10620-014-3473-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/27/2014] [Indexed: 12/12/2022]
Abstract
Autoimmune hepatitis is associated with interactive cell populations of the innate and adaptive immune systems, and these populations are amenable to therapeutic manipulation. The goals of this review are to describe the key cell populations implicated in autoimmune hepatitis and to identify investigational opportunities to develop cell-directed therapies for this disease. Studies cited in PubMed from 1972 to 2014 for autoimmune hepatitis, innate and adaptive immune systems, and therapeutic interventions were examined. Dendritic cells can promote immune tolerance to self-antigens, present neo-antigens that enhance the immune response, and expand the regulatory T cell population. Natural killer cells can secrete pro-inflammatory and anti-inflammatory cytokines and modulate the activity of dendritic cells and antigen-specific T lymphocytes. T helper 2 lymphocytes can inhibit the cytotoxic activities of T helper 1 lymphocytes and limit the expansion of T helper 17 lymphocytes. T helper 17 lymphocytes can promote inflammatory activity, and they can also up-regulate genes that protect against oxidative stress and hepatocyte apoptosis. Natural killer T cells can expand the regulatory T cell population; gamma delta lymphocytes can secrete interleukin-10, stimulate hepatic regeneration, and induce the apoptosis of hepatic stellate cells; and antigen-specific regulatory T cells can dampen immune cell proliferation and function. Pharmacological agents, neutralizing antibodies, and especially the adoptive transfer of antigen-specific regulatory T cells that have been freshly generated ex vivo are evolving as management strategies. The cells within the innate and adaptive immune systems are key contributors to the occurrence of autoimmune hepatitis, and they are attractive therapeutic targets.
Collapse
Affiliation(s)
- Aldo J Montano-Loza
- Division of Gastroenterology and Liver Unit, University of Alberta Hospital, Edmonton, AB, Canada
| | | |
Collapse
|
222
|
Generation of Alloreactive-Anergized Tr1 Cells From Patients on Dialysis for the Induction of Renal Transplant Tolerance: Are We There Yet? Transplantation 2015; 99:1551-2. [PMID: 26018353 DOI: 10.1097/tp.0000000000000752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
223
|
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
|
224
|
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
|
225
|
Abstract
PURPOSE OF REVIEW T regulatory cells (Tregs) play a central role in maintaining immune homeostasis and peripheral tolerance to foreign antigens in humans. The immune response to alloantigens and recurrence of autoimmunity contribute to pancreatic islet transplant dysfunction, hence the adoptive transfer of Tregs has the potential to significantly improve islet graft survival. In this review, we provide an in-depth analysis of challenges associated with the application of ex-vivo expanded Tregs therapy in pancreatic islet transplant. RECENT FINDINGS Tregs administered systemically may poorly migrate to the site of transplantation, which is critical for tolerance induction and graft protection. Intraportal administration of pancreatic tissue exerts some limitations on the ability to cotransplant Tregs at the same site of islet transplantation. In order to maximize therapeutic potential of Tregs, islet transplantation protocols may need additional refinement. Further to this, the Tregs may require cryopreservation in order to make them readily available at the same time as islet transplant. SUMMARY On the basis of current experience and technology, the combination of islet and Treg cotransplantation is feasible and has great potential to improve islet graft survival. The possibility to wean off, or withdraw, traditional immunosuppressive agents and improve patient quality of life makes it an interesting avenue to be pursued.
Collapse
|
226
|
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
|
227
|
Sicard A, Koenig A, Morelon E, Defrance T, Thaunat O. Cell therapy to induce allograft tolerance: time to switch to plan B? Front Immunol 2015; 6:149. [PMID: 25904913 PMCID: PMC4387960 DOI: 10.3389/fimmu.2015.00149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/19/2015] [Indexed: 12/13/2022] Open
Abstract
Organ transplantation is widely acknowledged as the best option for end stage failure of vital organs. Long-term graft survival is however limited by graft rejection, a destructive process resulting from the response of recipient’s immune system against donor-specific alloantigens. Prevention of rejection currently relies exclusively on immunosuppressive drugs that lack antigen specificity and therefore increase the risk for infections and cancers. Induction of donor-specific tolerance would provide indefinite graft survival without morbidity and therefore represents the grail of transplant immunologists. Progresses in the comprehension of immunoregulatory mechanisms over the last decades have paved the way for cell therapies to induce allograft tolerance. The first part of the present article reviews the promising results obtained in experimental models with adoptive transfer of ex vivo-expanded regulatory CD4+ T cells (CD4+ Tregs) and discuss which source and specificity should be preferred for transferred CD4+ Tregs. Interestingly, B cells have recently emerged as potent regulatory cells, able to establish a privileged crosstalk with CD4+ T cells. The second part of the present article reviews the evidences demonstrating the crucial role of regulatory B cells in transplantation tolerance. We propose the possibility to harness B cell regulatory functions to improve cell-based therapies aiming at inducing allograft tolerance.
Collapse
Affiliation(s)
| | - Alice Koenig
- U1111, INSERM , Lyon , France ; Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie Clinique , Lyon , France
| | - Emmanuel Morelon
- U1111, INSERM , Lyon , France ; Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie Clinique , Lyon , France ; Université de Lyon , Lyon , France
| | | | - Olivier Thaunat
- U1111, INSERM , Lyon , France ; Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie Clinique , Lyon , France ; Université de Lyon , Lyon , France
| |
Collapse
|
228
|
Abstract
There is a clear need to develop strategies to induce tolerance without the need of chronic immunosuppression in transplant recipient and in patients with autoimmunity. Adoptive T regulatory cell (Treg) therapy offers the potential of long-lasting protection. However, based on results of clinical trials so far with ex vivo expanded autologous Tregs in type 1 diabetic (T1D) patients, it seems unlikely that single immunotherapy with Treg infusion without immunomodulation regimens that promote stable donor Treg engraftment and persistence would afford truly significant clinical benefit. Combination therapies could provide improved outcomes with consideration of the fundamental factors required for Treg generation, homeostasis, and function to promote long-term donor Treg persistence to provoke beneficial therapeutic outcomes.
Collapse
|
229
|
New humanized mouse model of bronchiolitis obliterans syndrome. Transplantation 2015; 99:468-9. [PMID: 25695785 DOI: 10.1097/tp.0000000000000631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Humanized animals are transplanted with human tissues and cells to study their behavior as they do in the human body. This commentary briefly summarizes the recent developments and discusses the limitations of these humanized animal models.
Collapse
|
230
|
Ferrer IR, Hester J, Bushell A, Wood KJ. Induction of transplantation tolerance through regulatory cells: from mice to men. Immunol Rev 2015; 258:102-16. [PMID: 24517428 DOI: 10.1111/imr.12158] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Organ transplantation results in the activation of both innate and adaptive immune responses to the foreign antigens. While these responses can be limited with the use of systemic immunosuppressants, the induction of regulatory cell populations may be a novel strategy for the maintenance of specific immunological unresponsiveness that can reduce the severity of the detrimental side effects of current therapies. Our group has extensively researched different regulatory T-cell induction protocols for use as cellular therapy in transplantation. In this review, we address the cellular and molecular mechanisms behind regulatory T-cell suppression and their stability following induction protocols. We further discuss the use of different hematopoietically derived regulatory cell populations, including regulatory B cells, regulatory macrophages, tolerogenic dendritic cells, and myeloid-derived suppressor cells, for the induction of transplantation tolerance in light of new clinical trials developing therapies with some of these populations.
Collapse
Affiliation(s)
- Ivana R Ferrer
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
231
|
Jin X, Lu Y, Zhao Y, Yi S. Large-scale in vitro expansion of human regulatory T cells with potent xenoantigen-specific suppression. Cytotechnology 2015; 68:935-45. [PMID: 25605448 DOI: 10.1007/s10616-015-9845-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 01/13/2015] [Indexed: 02/05/2023] Open
Abstract
Xenotransplantation is a potential solution to the organ donor shortage. Immunosuppression is required for successful application of xenotransplantation but may lead to infection and cancer. Thus, strategies for immune tolerance induction need to be developed. Polyclonal regulatory T cells (Treg) play a central role in the induction and maintenance of immune tolerance and have been shown to protect against islet xenograft rejection in vivo. However, global immune suppression may be mediated by polyclonal Treg immunotherapy and a simple method for in vitro expansion of xenoantigen-specific Treg for efficient Treg application becomes necessary. Human Treg isolated from peripheral blood mononuclear cells (PBMCs) were initially cultured with anti-CD3/CD28 beads, rapamycin and IL-2 for 7 days as polyclonal expansion. Expanded Treg were then cocultured with irradiated porcine PBMC as xenoantigen stimulation for three subsequent cycles with 7 days for each cycle in the presence of IL-2 and anti-CD3/CD28 beads. Treg phenotype and suppressive capacity were assessed after each cycle of xenoantigen stimulation. Treg expanded with one cycle of xenoantigen stimulation retained Treg suppressive phenotype but acquired no xenoantigen specificity along with poor expansion efficiency, whereas expansion with two-cycle xenoantigen stimulation resulted in not only more than 800-fold Treg expansion but highly suppressive xenoantigen-specific Treg with effector Treg phenotype. However further increase of stimulation cycles resulted in reduced Treg suppressive potency. This study provides a simple approach to obtain high numbers of xenoantigen-specific Treg for immune tolerance induction in xenotransplantation.
Collapse
Affiliation(s)
- Xi Jin
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610000, Sichuan, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610000, Sichuan, China
- Center for Transplant and Renal Research, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, 610000, Sichuan, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610000, Sichuan, China
| | - Ye Zhao
- Center for Transplant and Renal Research, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Shounan Yi
- Center for Transplant and Renal Research, Westmead Hospital, Westmead, NSW, 2145, Australia.
- Transplantation Research Center, The 2nd Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
232
|
Pilon CB, Petillon S, Naserian S, Martin GH, Badoual C, Lang P, Azoulay D, Piaggio E, Grimbert P, Cohen JL. Administration of low doses of IL-2 combined to rapamycin promotes allogeneic skin graft survival in mice. Am J Transplant 2014; 14:2874-82. [PMID: 25394722 DOI: 10.1111/ajt.12944] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/25/2014] [Accepted: 07/12/2014] [Indexed: 01/25/2023]
Abstract
Human CD4(+) CD25(+) FoxP3(+) regulatory T cells (Tregs) prevent allogeneic graft rejection by inhibiting T cell activation, as has been shown in mouse models. Recently, low-dose IL-2 administration was shown to specifically activate Tregs but not pathogenic conventional T cells, leading to resolution of type 1 diabetes in nonobese diabetic mice. We therefore tested the ability of low-dose IL-2 to prevent allogeneic skin graft rejection. We found that while IL-2 alone was inefficient in preventing rejection, combined with rapamycin, IL-2 treatment promoted skin graft survival both in minor disparate and semi-allogeneic skin graft combinations. Tregs are activated by this combined treatment while conventional CD4(+) cell expansion and activation are markedly inhibited. Co-administration of anti-CD25 antibodies dramatically reduces the effect of the IL-2/rapamycin treatment, strongly supporting a central role for Treg activation. Thus, we provide the first preclinical data showing that low-dose IL-2 combined with rapamycin can significantly delay transplant rejection in mice. These findings may form the rational for clinical evaluation of this novel approach for the prevention of transplant rejection.
Collapse
Affiliation(s)
- C B Pilon
- Faculté de Médecine, Université Paris-Est Créteil, Créteil, France; Institut Mondor de Recherche Biomédicale (IMRB), INSERM U 955, Créteil, France; AP-HP, Groupe Hospitalier Henri-Mondor Albert-Chenevier, CIC-BT-504, Créteil, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Agarwal A, Fanelli G, Letizia M, Tung SL, Boardman D, Lechler R, Lombardi G, Smyth LA. Regulatory T cell-derived exosomes: possible therapeutic and diagnostic tools in transplantation. Front Immunol 2014; 5:555. [PMID: 25414702 PMCID: PMC4220709 DOI: 10.3389/fimmu.2014.00555] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/20/2014] [Indexed: 01/03/2023] Open
Abstract
Exosomes are extracellular vesicles released by many cells of the body. These small vesicles play an important part in intercellular communication both in the local environment and systemically, facilitating in the transfer of proteins, cytokines as well as miRNA between cells. The observation that exosomes isolated from immune cells such as dendritic cells (DCs) modulate the immune response has paved the way for these structures to be considered as potential immunotherapeutic reagents. Indeed, clinical trials using DC derived exosomes to facilitate immune responses to specific cancer antigens are now underway. Exosomes can also have a negative effect on the immune response and exosomes isolated from regulatory T cells (Tregs) and other subsets of T cells have been shown to have immune suppressive capacities. Here, we review what is currently known about Treg derived exosomes and their contribution to immune regulation, as well as highlighting their possible therapeutic potential for preventing graft rejection, and use as diagnostic tools to assess transplant outcome.
Collapse
Affiliation(s)
- Akansha Agarwal
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Giorgia Fanelli
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Marilena Letizia
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Sim Lai Tung
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Dominic Boardman
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Robert Lechler
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Giovanna Lombardi
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Lesley A Smyth
- Medical Research Council (MRC) Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| |
Collapse
|
234
|
Regulatory T-cell therapy in the induction of transplant tolerance: the issue of subpopulations. Transplantation 2014; 98:370-9. [PMID: 24933458 DOI: 10.1097/tp.0000000000000243] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Clinical tolerance induction to permit minimization or cessation of immunosuppressive drugs is one of the key research goals in solid organ transplantation. The use of ex vivo expanded or manipulated immunologic cells, including CD4CD25FOXP3 regulatory T cells (Tregs), to achieve this aim is already a reality, with several trials currently recruiting patients. Tregs are a highly suppressive, nonredundant, population of regulatory cells that prevent the development of autoimmune diseases in mammals. Data from transplanted humans and animal models support the notion that Tregs can mediate both induction and adoptive transfer of transplantation tolerance. However, human Tregs are highly heterogeneous and include subpopulations with the potential to produce the proinflammatory cytokine interleukin-17, which has been linked to transplant rejection. Tregs are also small in number in the peripheral circulation, thus they require ex vivo expansion before infusion into man. Selection of the most appropriate Treg population for cell therapy is, therefore, a critical step in ensuring successful clinical outcomes. In this review, we discuss Treg subpopulations, their subdivision based on nonmutually exclusive criteria of origin, expression of immunologic markers and function, availability in the peripheral blood of patients awaiting transplantation, and their suitability for programs of cell-based therapy.
Collapse
|
235
|
Immunosuppressive therapy in allograft transplantation: from novel insights and strategies to tolerance and challenges. Cent Eur J Immunol 2014; 39:400-9. [PMID: 26155155 PMCID: PMC4440012 DOI: 10.5114/ceji.2014.45955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 07/03/2014] [Indexed: 01/07/2023] Open
Abstract
Immunosuppression therapy is the key to successful post-transplantation outcomes. The need for ideal immunosuppression became durable maintenance of long-term graft survival. In spite of current immunosuppressive therapy regimens advances, surgical procedures, and preservation methods, organ transplantation is associated with a long-term poor survival and significant mortality. This has led to an increased interest to optimize outcomes while minimizing associated toxicity by using alternative methods for maintenance immunosuppression, organ rejection treatment, and monitoring of immunosuppression. T regulatory (Treg) cells, which have immunosuppressive functions and cytokine profiles, have been studied during the last decades. Treg cells are able to inhibit the development of allergen-specific cell responses and consequently play a key role in a healthy immune response to allergens. Mature dendritic cells (DCs) play a crucial role in the differentiation of Tregs, which are known to regulate allergic inflammatory responses. Advance in long-standing allograft outcomes may depend on new drugs with novel mechanisms of action with minimal toxicity. Newer treatment techniques have been developed, including using novel stem cell-based therapies such as mesenchymal stem cells, phagosomes and exosomes. Immunoisolation techniques and salvage therapies, including photopheresis and total lymphoid irradiation have emerged as alternative therapeutic choices. The present review evaluates the recent clinical advances in immunosuppressive therapies for organ transplantation.
Collapse
|
236
|
Cohen JL, Pilon C, Maury S, Grimbert P. Searching for factors to improve regulatory T cell therapy in organ transplantation. Am J Transplant 2014; 14:2430-1. [PMID: 25039852 DOI: 10.1111/ajt.12855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- J L Cohen
- Faculté de médecine, Université Paris-Est Créteil, Créteil, France; INSERM U 955, Institut Mondor de Recherche Biomédicale, Créteil, France; AP-HP, Groupe Hospitalier Henri-Mondor Albert-Chenevier, Créteil, France
| | | | | | | |
Collapse
|
237
|
Noyan F, Lee YS, Zimmermann K, Hardtke-Wolenski M, Taubert R, Warnecke G, Knoefel AK, Schulde E, Olek S, Manns MP, Jaeckel E. Isolation of human antigen-specific regulatory T cells with high suppressive function. Eur J Immunol 2014; 44:2592-602. [DOI: 10.1002/eji.201344381] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/10/2014] [Accepted: 06/30/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Fatih Noyan
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | - Young-Seon Lee
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | - Katharina Zimmermann
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | - Gregor Warnecke
- Department of Cardiothoracic, Transplantation and Vascular Surgery; Hannover Medical School; Hannover Germany
| | - Ann-Kathrin Knoefel
- Department of Cardiothoracic, Transplantation and Vascular Surgery; Hannover Medical School; Hannover Germany
| | - Elvira Schulde
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | | | - Michael P. Manns
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology & Endocrinology; Hannover Medical School; Hannover Germany
| |
Collapse
|
238
|
Govender L, Pascual M, Golshayan D. Potential and limitations of regulatory T-cell therapy in solid organ transplantation. Expert Rev Clin Immunol 2014; 10:1197-212. [PMID: 25073810 DOI: 10.1586/1744666x.2014.943191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Over the past few years, the therapeutic potential of Treg has been highlighted in the field of autoimmune diseases and after allogeneic transplantation. The first hurdle for the therapeutic use of Treg is their insufficient numbers in non-manipulated individuals, in particular when facing strong immune activation and expanding effector cells, such as in response to an allograft. Here we review current approaches being explored for Treg expansion in the perspective of clinical therapeutic protocols. We describe different Treg subsets that could be suitable for clinical application, as well as discuss factors such as the required dose of Treg, their antigen-specificity and in vivo stability, that have to be considered for optimal Treg-based immunotherapy in transplantation. Since Treg may not be sufficient as stand-alone therapy for solid organ transplantation in humans, we draw attention to possible hurdles and combination therapy with immunomodulatory drugs that could possibly improve the in vivo efficacy of Treg.
Collapse
Affiliation(s)
- Lerisa Govender
- Departments of Medicine and Surgery, Transplantation Centre and Transplantation Immunopathology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, 1011 Lausanne, Switzerland
| | | | | |
Collapse
|
239
|
Kanakry CG, Ganguly S, Zahurak M, Bolaños-Meade J, Thoburn C, Perkins B, Fuchs EJ, Jones RJ, Hess AD, Luznik L. Aldehyde dehydrogenase expression drives human regulatory T cell resistance to posttransplantation cyclophosphamide. Sci Transl Med 2014; 5:211ra157. [PMID: 24225944 DOI: 10.1126/scitranslmed.3006960] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
High-dose, posttransplantation cyclophosphamide (PTCy) is an effective strategy for preventing graft-versus-host disease (GVHD) after allogeneic blood or marrow transplantation (alloBMT). However, the mechanisms by which PTCy modulates alloimmune responses are not well understood. We studied early T cell reconstitution in patients undergoing alloBMT with PTCy and the effects of mafosfamide, a cyclophosphamide (Cy) analog, on CD4(+) T cells in allogeneic mixed lymphocyte reactions (MLRs) in vitro. Patients exhibited reductions in naïve, potentially alloreactive conventional CD4(+) T cells with relative preservation of memory CD4(+)Foxp3(+) T cells. In particular, CD4(+)CD45RA(-)Foxp3(+hi) effector regulatory T cells (Tregs) recovered rapidly after alloBMT and, unexpectedly, were present at higher levels in patients with GVHD. CD4(+)Foxp3(+) T cells from patients and from allogeneic MLRs expressed relatively high levels of aldehyde dehydrogenase (ALDH), the major in vivo mechanism of Cy resistance. Treatment of MLR cultures with the ALDH inhibitor diethylaminobenzaldehyde reduced the activation and proliferation of CD4(+) T cells and sensitized Tregs to mafosfamide. Finally, removing Tregs from peripheral blood lymphocyte grafts obviated PTCy's GVHD-protective effect in a xenogeneic transplant model. Together, these findings suggest that Treg resistance to Cy through expression of ALDH may contribute to the clinical activity of PTCy in preventing GVHD.
Collapse
Affiliation(s)
- Christopher G Kanakry
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Cortés JR, Sánchez-Díaz R, Bovolenta ER, Barreiro O, Lasarte S, Matesanz-Marín A, Toribio ML, Sánchez-Madrid F, Martín P. Maintenance of immune tolerance by Foxp3+ regulatory T cells requires CD69 expression. J Autoimmun 2014; 55:51-62. [PMID: 24934597 DOI: 10.1016/j.jaut.2014.05.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/22/2014] [Accepted: 05/23/2014] [Indexed: 11/24/2022]
Abstract
Although FoxP3(+) regulatory T cells are key players in the maintenance of immune tolerance and autoimmunity, the lack of specific markers constitute an obstacle to their use for immunotherapy protocols. In this study, we have investigated the role of the C-type lectin receptor CD69 in the suppressor function of Tregs and maintenance of immune tolerance towards harmless inhaled antigens. We identified a novel FoxP3(+)CD69(+) Treg subset capable to maintain immune tolerance and protect to developing inflammation. Although CD69(+) and CD69(-)FoxP3(+) Tregs exist in homeostasis, only CD69-expressing Tregs express high levels of CTLA-4, ICOS, CD38 and GITR suppression-associated markers, secrete high amounts of TGFβ and have potent suppressor activity. This activity is regulated by STAT5 and ERK signaling pathways and is impaired by antibody-mediated down-regulation of CD69 expression. Moreover, immunotherapy with FoxP3(+)CD69(+) Tregs restores the homeostasis in Cd69(-/-) mice, that fail to induce tolerance, and is also highly proficient in the prevention of inflammation. The identification of the FoxP3(+)CD69(+) Treg subset paves the way toward the development of new therapeutic strategies to control immune homeostasis and autoimmunity.
Collapse
Affiliation(s)
- José R Cortés
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| | - Raquel Sánchez-Díaz
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| | - Elena R Bovolenta
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| | - Olga Barreiro
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| | - Sandra Lasarte
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| | - Adela Matesanz-Marín
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| | - María L Toribio
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Spain
| | - Francisco Sánchez-Madrid
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain.,Servicio de Inmunología, Hospital de La Princesa, Universidad Autónoma de Madrid, Madrid, 28006 Spain
| | - Pilar Martín
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029 Spain
| |
Collapse
|
241
|
Picarda E, Bézie S, Venturi V, Echasserieau K, Mérieau E, Delhumeau A, Renaudin K, Brouard S, Bernardeau K, Anegon I, Guillonneau C. MHC-derived allopeptide activates TCR-biased CD8+ Tregs and suppresses organ rejection. J Clin Invest 2014; 124:2497-512. [PMID: 24789907 DOI: 10.1172/jci71533] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In a rat heart allograft model, preventing T cell costimulation with CD40Ig leads to indefinite allograft survival, which is mediated by the induction of CD8+CD45RClo regulatory T cells (CD8+CD40Ig Tregs) interacting with plasmacytoid dendritic cells (pDCs). The role of TCR-MHC-peptide interaction in regulating Treg activity remains a topic of debate. Here, we identified a donor MHC class II-derived peptide (Du51) that is recognized by TCR-biased CD8+CD40Ig Tregs and activating CD8+CD40Ig Tregs in both its phenotype and suppression of antidonor alloreactive T cell responses. We generated a labeled tetramer (MHC-I RT1.Aa/Du51) to localize and quantify Du51-specific T cells within rat cardiac allografts and spleen. RT1.Aa/Du51-specific CD8+CD40Ig Tregs were the most suppressive subset of the total Treg population, were essential for in vivo tolerance induction, and expressed a biased, restricted Vβ11-TCR repertoire in the spleen and the graft. Finally, we demonstrated that treatment of transplant recipients with the Du51 peptide resulted in indefinite prolongation of allograft survival. These results show that CD8+CD40Ig Tregs recognize a dominant donor antigen, resulting in TCR repertoire alterations in the graft and periphery. Furthermore, this allopeptide has strong therapeutic activity and highlights the importance of TCR-peptide-MHC interaction for Treg generation and function.
Collapse
|
242
|
Seddiki N, Cook L, Hsu DC, Phetsouphanh C, Brown K, Xu Y, Kerr SJ, Cooper DA, Munier CML, Pett S, Ananworanich J, Zaunders J, Kelleher AD. Human antigen-specific CD4+CD25+CD134+CD39+T cells are enriched for regulatory T cells and comprise a substantial proportion of recall responses. Eur J Immunol 2014; 44:1644-61. [DOI: 10.1002/eji.201344102] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 01/28/2014] [Accepted: 03/12/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Nabila Seddiki
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Laura Cook
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Denise C. Hsu
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV Netherlands Australia Thailand Research Collaboration; Thai Red Cross AIDS Research Centre; Bangkok Thailand
| | - Chansavath Phetsouphanh
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Kai Brown
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Yin Xu
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Stephen J. Kerr
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV Netherlands Australia Thailand Research Collaboration; Thai Red Cross AIDS Research Centre; Bangkok Thailand
| | - David A. Cooper
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV; Immunology and Infectious Diseases Clinical Services Unit; St Vincent's Hospital; Sydney NSW Australia
| | - C. Mee Ling Munier
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
| | - Sarah Pett
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV; Immunology and Infectious Diseases Clinical Services Unit; St Vincent's Hospital; Sydney NSW Australia
| | - Jintanat Ananworanich
- HIV Netherlands Australia Thailand Research Collaboration; Thai Red Cross AIDS Research Centre; Bangkok Thailand
| | - John Zaunders
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
| | - Anthony D. Kelleher
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV; Immunology and Infectious Diseases Clinical Services Unit; St Vincent's Hospital; Sydney NSW Australia
| |
Collapse
|
243
|
Juvet SC, Whatcott AG, Bushell AR, Wood KJ. Harnessing regulatory T cells for clinical use in transplantation: the end of the beginning. Am J Transplant 2014; 14:750-63. [PMID: 24592900 DOI: 10.1111/ajt.12647] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/19/2013] [Accepted: 12/19/2013] [Indexed: 01/25/2023]
Abstract
Owing to the adverse effects of immunosuppression and an inability to prevent chronic rejection, there is a pressing need for alternative strategies to control alloimmunity. In three decades, regulatory T cells (Tregs) have evolved from a hypothetical mediator of adoptively transferred tolerance to a well-defined population that can be expanded ex vivo and returned safely to patients in clinical trials. Herein, we review the historical developments that have permitted these advances and the current status of clinical trials examining Tregs as a cellular therapy in transplantation. We conclude by discussing the critical unanswered questions that face this field in the coming years.
Collapse
Affiliation(s)
- S C Juvet
- Nuffield Department of Surgical Sciences, Transplantation Research Immunology Group, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | |
Collapse
|
244
|
Waldmann H, Hilbrands R, Howie D, Cobbold S. Harnessing FOXP3+ regulatory T cells for transplantation tolerance. J Clin Invest 2014; 124:1439-45. [PMID: 24691478 DOI: 10.1172/jci67226] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Early demonstrations that mice could be tolerized to transplanted tissues with short courses of immunosuppressive therapy and that with regard to tolerance to self, CD4+FOXP3+ regulatory T cells (Tregs) appeared to play a critical role, have catalyzed strategies to harness FOXP3-dependent processes to control rejection in human transplantation. This review seeks to examine the scientific underpinning for this new approach to finesse immunosuppression.
Collapse
|
245
|
Xiao F, Ma L, Zhao M, Huang G, Mirenda V, Dorling A, Lechler R, Lombardi G. Ex vivo expanded human regulatory T cells delay islet allograft rejection via inhibiting islet-derived monocyte chemoattractant protein-1 production in CD34+ stem cells-reconstituted NOD-scid IL2rγnull mice. PLoS One 2014; 9:e90387. [PMID: 24594640 PMCID: PMC3940883 DOI: 10.1371/journal.pone.0090387] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/28/2014] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by immune-mediated destruction of insulin-secreting β cells of the pancreas. Near complete dependence on exogenous insulin makes T1DM very difficult to control, with the result that patients are exposed to high blood glucose and risk of diabetic complications and/or intermittent low blood glucose that can cause unconsciousness, fits and even death. Allograft transplantation of pancreatic islets restores normoglycemia with a low risk of surgical complications. However, although successful immediately after transplantation, islets are progressively lost, with most of the patients requiring exogenous insulin within 2 years post-transplant. Therefore, there is an urgent requirement for the development of new strategies to prevent islet rejection. In this study, we explored the importance of human regulatory T cells in the control of islets allograft rejection. We developed a pre-clinical model of human islet transplantation by reconstituting NOD-scid IL2rγnull mice with cord blood-derived human CD34+ stem cells and demonstrated that although the engrafted human immune system mediated the rejection of human islets, their survival was significantly prolonged following adoptive transfer of ex vivo expanded human Tregs. Mechanistically, Tregs inhibited the infiltration of innate immune cells and CD4+ T cells into the graft by down-regulating the islet graft-derived monocyte chemoattractant protein-1. Our findings might contribute to the development of clinical strategies for Treg therapy to control human islet rejection. We also show for the first time that CD34+ cells-reconstituted NOD-scid IL2rγnull mouse model could be beneficial for investigating human innate immunity in vivo.
Collapse
Affiliation(s)
- Fang Xiao
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Liang Ma
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Min Zhao
- Department of Diabetes & Endocrinology, King's College London, London, United Kingdom
| | - Guocai Huang
- Department of Diabetes & Endocrinology, King's College London, London, United Kingdom
| | - Vincenzo Mirenda
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Anthony Dorling
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Robert Lechler
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
246
|
Vingert B, Tamagne M, Desmarets M, Pakdaman S, Elayeb R, Habibi A, Bernaudin F, Galacteros F, Bierling P, Noizat-Pirenne F, Cohen J, Cohen J. Partial dysfunction of Treg activation in sickle cell disease. Am J Hematol 2014; 89:261-6. [PMID: 24779034 DOI: 10.1002/ajh.23629] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Sickle cell disease (SCD) is a chronic inflammatory disease associated with multiple organ damage, chronic anemia, and infections. SCD patients have a high rate of alloimmunization against red blood cells (RBCs) following transfusion and may develop autoimmune diseases. Studies in mouse models have suggested that regulatory T cells (Treg) play a role in alloimmunization against RBC antigens. We characterized the phenotype and function of the Treg cell population in a homogeneous cohort of transfused SCD patients. We found that the distribution of Treg subpopulations differed significantly between SCD patients and healthy blood donors. SCD patients have a particular Treg phenotype, with strong CTLA-4 and CD39 expression and weak HLA-DR and CCR7 expression. Finally, we show that this particular phenotype is related to SCD rather than alloimmunization status. Indeed, we observed no difference in Treg phenotype or function in vitro using autologous feeder cells between strong and weak responders to alloimmunization.
Collapse
Affiliation(s)
- Benoît Vingert
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
| | - Marie Tamagne
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
| | - Maxime Desmarets
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
| | - Sadaf Pakdaman
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
| | - Rahma Elayeb
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
| | - Anoosha Habibi
- AP-HP; Service Maladies Génétiques du Globule Rouge; Hôpital Henri Mondor 94200 Créteil France
| | - Françoise Bernaudin
- Centre de Référence de Drépanocytose-Centre Hospitalier Intercommunal de Créteil; 94200 Créteil France
| | - Frédéric Galacteros
- Inserm, U955; IMRB Equipe 2 Créteil France
- AP-HP; Service Maladies Génétiques du Globule Rouge; Hôpital Henri Mondor 94200 Créteil France
- Université Paris-Est, UPEC; Créteil France
| | - Philippe Bierling
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
| | - France Noizat-Pirenne
- EFS Île de France; Hôpital Henri Mondor; 94200 Créteil France
- Inserm, U955; IMRB Equipe 2 Créteil France
- Université Paris-Est, UPEC; Créteil France
| | - José Cohen
- Université Paris-Est, UPEC; Créteil France
- Inserm, U955; Equipe 21 Créteil France
- AP-HP; Hôpital Henri-Mondor - A. Chenevier; CIC-BT-504 Créteil France
| | | |
Collapse
|
247
|
Abstract
Tolerance induction and alloreactivity can be applied to the clinic for the transplantation of solid organs and in the treatment of human cancers respectively. Hematopoietic chimerism, the stable coexistence of host and donor blood cells, guarantees that a solid organ from the same donor will be tolerated without a requirement for maintenance immunosuppression, and it also serves as a platform for the adoptive immunotherapy of hematologic malignancies using donor lymphocyte infusions. This review focuses on clinically relevant methods for inducing hematopoietic chimerism and transplantation tolerance, with a special emphasis on reduced intensity transplantation conditioning and high dose, post-transplantation cyclophosphamide to prevent graft rejection and graft-versus-host disease (GVHD). Reduced intensity transplantation regimens permit a transient cooperation between donor and host immune systems to eradicate malignancy without producing GVHD. Their favorable toxicity profile also enables the application of allogeneic stem cell transplantation to treat non-malignant disorders of hematopoiesis and to induce tolerance for solid organ transplantation.
Collapse
Affiliation(s)
- Ephraim J. Fuchs
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| |
Collapse
|
248
|
Landwehr-Kenzel S, Issa F, Luu SH, Schmück M, Lei H, Zobel A, Thiel A, Babel N, Wood K, Volk HD, Reinke P. Novel GMP-compatible protocol employing an allogeneic B cell bank for clonal expansion of allospecific natural regulatory T cells. Am J Transplant 2014; 14:594-606. [PMID: 24467477 DOI: 10.1111/ajt.12629] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/13/2013] [Indexed: 01/25/2023]
Abstract
The adoptive transfer of natural regulatory T cells (nTreg) is a new option to reshape undesired immune reactivity in autoimmunity and transplantation toward "tolerance." The first clinical trials using adoptive transfer of polyclonal nTreg demonstrated safety and hints of efficacy. However, the low frequencies of antigen-specific cells among the pool of polyclonal nTreg and their broad antigen nonspecific suppression are limitations of this approach regarding efficacy and safety. Recently, the isolation and expansion of (allo)antigen-specific nTreg have successfully been achieved by using Treg-specific activation markers but the yield is relatively low. Here, we describe a novel good manufacturing practice (GMP)-compatible expansion protocol of alloantigen-specific nTreg based on the stimulation of nTreg by allogeneic activated B cells. Their functionality and specificity are superior compared to polyclonal nTreg both in vitro and in vivo. Employing an allogeneic B cell bank, designed to cover the majority of HLA types, allows fast GMP-compliant manufacturing for donor-specific nTreg for clinical application in organ and stem cell transplantation. TCR repertoire analyses by next generation sequencing revealed impressive expansion by several log-steps of even very low-abundance alloantigen-specific nTreg clones. This novel method offers a simple approach for expanding antigen-specific nTreg and is characterized by high replicability and easy transferability to full GMP standards.
Collapse
Affiliation(s)
- S Landwehr-Kenzel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany; Department of Pediatric Pulmonology and Immunology, Charité University Medicine Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Imanguli MM, Cowen EW, Rose J, Dhamala S, Swaim W, Lafond S, Yagi B, Gress RE, Pavletic SZ, Hakim FT. Comparative analysis of FoxP3(+) regulatory T cells in the target tissues and blood in chronic graft versus host disease. Leukemia 2014; 28:2016-27. [PMID: 24577531 DOI: 10.1038/leu.2014.92] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 02/09/2014] [Accepted: 02/18/2014] [Indexed: 01/11/2023]
Abstract
Activation and migration of regulatory T cells (Treg) into tissue is critical in control of inflammation, but has not been examined extensively in chronic graft versus host disease (cGVHD). In parallel studies of tissues and blood, we determined that FoxP3(+) T cells increased in proportion to T effectors (Teff) in tissue infiltrates in oral and cutaneous lichenoid cGVHD. These FoxP3(+) cells expressed distinguishing phenotypic and functional markers of Treg (CD3(+), CD4(+), CD27(+), ICOS(+) and CD39(+)), not found on FoxP3(-) Teff. Both Teff and FoxP3(+) Treg expressed T-bet and the chemokine receptor CXCR3, however, consistent with a common mechanism of chemokine-mediated migration into tissue. Furthermore, functional markers (ICOS and CD39) and chemokine receptors (CXCR3) were both present in a higher proportion of FoxP3(+) cells in tissues than in peripheral blood, consistent with recruitment and activation of Treg in cGVHD target tissues. Finally, the 'activated' CD45RA(-)FoxP3(hi) subset of Treg cells, which highly express functional markers, were found in comparable frequencies in cGVHD patients and normal controls, despite a significant deficit in naive 'resting' Treg. These findings are consistent with Treg capacity to upregulate functional markers and traffick into tissue in cGVHD.
Collapse
Affiliation(s)
- M M Imanguli
- 1] Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA [2] Department of Otolaryngology Head and Neck Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - E W Cowen
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - J Rose
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - S Dhamala
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - W Swaim
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - S Lafond
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - B Yagi
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - R E Gress
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - S Z Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - F T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
250
|
Retinoic acid stabilizes antigen-specific regulatory T-cell function in autoimmune hepatitis type 2. J Autoimmun 2014; 53:26-32. [PMID: 24566085 DOI: 10.1016/j.jaut.2014.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 02/02/2014] [Indexed: 01/03/2023]
Abstract
Imbalance between effector and regulatory T-cells (Treg) underlies the loss of immune-tolerance to self-antigens in autoimmune disease. In autoimmune hepatitis type 2 (AIH-2), effector CD4 T-cell immune responses to cytochrome P450IID6 (CYP2D6) are permitted by numerically and functionally impaired Treg. Restoration of CYP2D6-specific Treg in AIH-2 would enable control over effectors sharing the same antigen specificity, leading to re-establishment of immune-tolerance. We have previously developed a protocol for generating antigen-specific Treg through co-culture with semi-mature dendritic cells presenting CYP2D6 peptides. In this study, we aimed to explore phenotypic and functional features of patient Treg compared to health, to test Treg stability under pro-inflammatory conditions, and to investigate the potential benefit of supplementation with all-trans-retinoic acid (RA) or rapamycin (RP), agents proven to enhance Treg function. We show that antigen-specific Treg from patients have comparable phenotypic and functional features to those from healthy controls, suppressing both proliferation and pro-inflammatory cytokine production by effector cells. Treg exposure to inflammatory challenge results in decreased suppressive function and up-regulation of Th1/Th2/Th17 transcription factors both in health and AIH-2. The increase of Th1 and Th17 transcription factors is limited by addition of RA in controls and Th1 expression is decreased by RP in patients. Importantly, inflammation-induced decrease in Treg function is also abrogated by RA/RP in health and RA in patients. Our data provide important information for the optimization of protocols aiming at generating antigen-specific Treg for treatment of autoimmune disease and for understanding their biology upon pro-inflammatory challenge and RP/RA supplementation.
Collapse
|