1
|
Jang C, Hsu J. Allogeneic Hematopoietic Stem Cell Transplantation After Solid Organ Transplantation in Patients With Hematologic Malignancies Managed With Post-Transplant Cyclophosphamide-Based Graft-Versus-Host Disease Prophylaxis. J Hematol 2024; 13:250-258. [PMID: 39493605 PMCID: PMC11526586 DOI: 10.14740/jh1327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/19/2024] [Indexed: 11/05/2024] Open
Abstract
Patients who receive solid organ transplants often require lifelong immunosuppression, which increases their risk for hematologic disorders. Allogeneic hematopoietic stem cell transplantation (HSCT) offers a potential curative treatment option for these patients. However, there is still a lack of understanding and guidance on graft-vs-host disease (GVHD) immunosuppression regimens, potential complications, and outcomes in patients with solid organ transplants who undergo HSCT. The rate of solid organ transplantation continues to increase annually, making this a common clinical scenario that hematologists encounter. In this case series, we present three patients who underwent liver, kidney and cardiac transplants and each developed hematological malignancies requiring allogeneic stem cell transplant. This is the first case report of two patients who received post-transplant cyclophosphamide with mycophenolate mofetil and tacrolimus GVHD prophylaxis. We also review recent advances in GVHD prophylaxis in allogeneic HSCT and solid organ transplantation including immune tolerance and immunosuppression-free protocols. Our case series support the use of post-transplant cyclophosphamide with mycophenolate mofetil and tacrolimus as post-transplant GVHD prophylaxis, which does not appear to compromise solid organ graft function. Our case series also provides evidence that allogeneic HSCT is a feasible and potentially life-saving treatment option in patients who develop hematologic malignancies after solid organ transplantation.
Collapse
Affiliation(s)
- Charley Jang
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Jingmei Hsu
- Department of Hematology and Oncology, NYU Langone Health Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
2
|
Saha I, Chawla AS, Oliveira APBN, Elfers EE, Warrick K, Meibers HE, Jain VG, Hagan T, Katz JD, Pasare C. Alloreactive memory CD4 T cells promote transplant rejection by engaging DCs to induce innate inflammation and CD8 T cell priming. Proc Natl Acad Sci U S A 2024; 121:e2401658121. [PMID: 39136987 PMCID: PMC11348247 DOI: 10.1073/pnas.2401658121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Alloreactive memory T cells have been implicated as central drivers of transplant rejection. Perplexingly, innate cytokines, such as IL-6, IL-1β, and IL-12, are also associated with rejection of organ transplants. However, the pathways of innate immune activation in allogeneic transplantation are unclear. While the role of microbial and cell death products has been previously described, we identified alloreactive memory CD4 T cells as the primary triggers of innate inflammation. Memory CD4 T cells engaged MHC II-mismatched dendritic cells (DCs), leading to the production of innate inflammatory cytokines. This innate inflammation was independent of several pattern recognition receptors and was primarily driven by TNF superfamily ligands expressed by alloreactive memory CD4 T cells. Blocking of CD40L and TNFα resulted in dampened inflammation, and mice genetically deficient in these molecules exhibited prolonged survival of cardiac allografts. Furthermore, myeloid cell and CD8 T cell infiltration into cardiac transplants was compromised in both CD40L- and TNFα-deficient recipients. Strikingly, we found that priming of naive alloreactive CD8 T cells was dependent on licensing of DCs by memory CD4 T cells. This study unravels the key mechanisms by which alloreactive memory CD4 T cells contribute to destructive pathology and transplant rejection.
Collapse
Affiliation(s)
- Irene Saha
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Amanpreet Singh Chawla
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Ana Paula B. N. Oliveira
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Eileen E. Elfers
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Kathrynne Warrick
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH45220
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Hannah E. Meibers
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH45220
| | - Viral G. Jain
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Thomas Hagan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH45220
| | - Jonathan D. Katz
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH45220
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH45220
| |
Collapse
|
3
|
Chaban R, Ileka I, McGrath G, Kinoshita K, Habibabady Z, Ma M, Diaz V, Maenaka A, Calhoun A, Dufault M, Rosales I, Laguerre CM, Sanatkar SA, Burdorf L, Ayares DL, Eyestone W, Sardana P, Kuravi K, Sorrells L, Lederman S, Lucas CG, Prather RS, Wells KD, Whitworth KM, Cooper DKC, Pierson RN. Extended survival of 9- and 10-gene-edited pig heart xenografts with ischemia minimization and CD154 costimulation blockade-based immunosuppression. J Heart Lung Transplant 2024:S1053-2498(24)01777-7. [PMID: 39097214 DOI: 10.1016/j.healun.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 07/26/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Xenotransplantation has made significant advances recently using pigs genetically engineered to remove carbohydrate antigens, either alone or with addition of various human complement, coagulation, and anti-inflammatory ''transgenes''. Here we evaluated results associated with gene-edited (GE) pig hearts transplanted in baboons using an established costimulation-based immunosuppressive regimen and a cold-perfused graft preservation technique. METHODS Eight baboons received heterotopic abdominal heart transplants from 3-GE (GalKO.β4GalNT2KO.hCD55, n = 3), 9-GE (GalKO.β4GalNT2KO.GHRKO.hCD46.hCD55. TBM.EPCR.hCD47. HO-1, n = 3) or 10-G (9-GE+CMAHKO, n = 2) pigs using Steen's cold continuous perfusion for ischemia minimization. Immunosuppression (IS) included induction with anti-thymocyte globulin and αCD20, ongoing αCD154, MMF, and tapered corticosteroid. RESULTS All three 3-GE grafts functioned well initially, but failed within 5 days. One 9-GE graft was lost intraoperatively due to a technical issue and another was lost at POD 13 due to antibody mediated rejection (AMR) in a baboon with a strongly positive pre-operative cross-match. One 10-GE heart failed at POD113 with combined cellular and antibody mediated rejection. One 9-GE and one 10-GE hearts had preserved graft function with normal myocardium on protocol biopsies, but exhibited slowly progressive graft hypertrophy until elective necropsy at POD393 and 243 respectively. Elevated levels of IL-6, MCP-1, C-reactive protein, and human thrombomodulin were variably associated with conditioning, the transplant procedure, and clinically significant postoperative events. CONCLUSION Relative to reference genetics without thrombo-regulatory and anti-inflammatory gene expression, 9- or 10-GE pig hearts exhibit promising performance in the context of a clinically applicable regimen including ischemia minimization and αCD154-based IS, justifying further evaluation in an orthotopic model.
Collapse
Affiliation(s)
- Ryan Chaban
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Department of Cardiovascular Surgery, University Hospital of Mainz, Mainz, Germany
| | - Ikechukwu Ileka
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gannon McGrath
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kohei Kinoshita
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zahra Habibabady
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Madelyn Ma
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Victoria Diaz
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Akihiro Maenaka
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anthony Calhoun
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Megan Dufault
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ivy Rosales
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Christiana M Laguerre
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Seyed-Amir Sanatkar
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lars Burdorf
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Revivicor, Inc., Blacksburg, Virginia
| | | | | | | | | | | | | | - Caroline G Lucas
- National Swine Resource and Research Center (NSRRC), Animal Science Research Center, University of Missouri, Columbia, Missouri
| | - Randall S Prather
- National Swine Resource and Research Center (NSRRC), Animal Science Research Center, University of Missouri, Columbia, Missouri
| | - Kevin D Wells
- National Swine Resource and Research Center (NSRRC), Animal Science Research Center, University of Missouri, Columbia, Missouri
| | - Kristin M Whitworth
- National Swine Resource and Research Center (NSRRC), Animal Science Research Center, University of Missouri, Columbia, Missouri
| | - David K C Cooper
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard N Pierson
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
4
|
Grosu-Bularda A, Hodea FV, Zamfirescu D, Stoian A, Teodoreanu RN, Lascăr I, Hariga CS. Exploring Costimulatory Blockade-Based Immunologic Strategies in Transplantation: Are They a Promising Immunomodulatory Approach for Organ and Vascularized Composite Allotransplantation? J Pers Med 2024; 14:322. [PMID: 38541064 PMCID: PMC10971463 DOI: 10.3390/jpm14030322] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 11/12/2024] Open
Abstract
The field of transplantation, including the specialized area of vascularized composite allotransplantation (VCA), has been transformed since the first hand transplant in 1998. The major challenge in VCA comes from the need for life-long immunosuppressive therapy due to its non-vital nature and a high rate of systemic complications. Ongoing research is focused on immunosuppressive therapeutic strategies to avoid toxicity and promote donor-specific tolerance. This includes studying the balance between tolerance and effector mechanisms in immune modulation, particularly the role of costimulatory signals in T lymphocyte activation. Costimulatory signals during T cell activation can have either stimulatory or inhibitory effects. Interfering with T cell activation through costimulation blockade strategies shows potential in avoiding rejection and prolonging the survival of transplanted organs. This review paper aims to summarize current data on the immunologic role of costimulatory blockade in the field of transplantation. It focuses on strategies that can be applied in vascularized composite allotransplantation, offering insights into novel methods for enhancing the success and safety of these procedures.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | | | | | - Răzvan Nicolae Teodoreanu
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | - Ioan Lascăr
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | - Cristian Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| |
Collapse
|
5
|
Mao K, Wang J, Xie Q, Yang YG, Shen S, Sun T, Wang J. Cationic nanoparticles-based approaches for immune tolerance induction in vivo. J Control Release 2024; 366:425-447. [PMID: 38154540 DOI: 10.1016/j.jconrel.2023.12.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/04/2023] [Accepted: 12/25/2023] [Indexed: 12/30/2023]
Abstract
The development of autoimmune diseases and the rejection of transplanted organs are primarily caused by an exaggerated immune response to autoantigens or graft antigens. Achieving immune tolerance is crucial for the effective treatment of these conditions. However, traditional therapies often have limited therapeutic efficacy and can result in systemic toxic effects. The emergence of nanomedicine offers a promising avenue for addressing immune-related diseases. Among the various nanoparticle formulations, cationic nanoparticles have demonstrated significant potential in inducing immune tolerance. In this review, we provide an overview of the underlying mechanism of autoimmune disease and organ transplantation rejection. We then highlight the recent advancements and advantages of utilizing cationic nanoparticles for inducing immune tolerance in the treatment of autoimmune diseases and the prevention of transplant rejection.
Collapse
Affiliation(s)
- Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Qianyue Xie
- Huafu International Department, Affiliated High School of South China Normal University, Guangzhou, Guangdong, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China; Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovatiion Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Ahuja HK, Azim S, Maluf D, Mas VR. Immune landscape of the kidney allograft in response to rejection. Clin Sci (Lond) 2023; 137:1823-1838. [PMID: 38126208 DOI: 10.1042/cs20230493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Preventing kidney graft dysfunction and rejection is a critical step in addressing the nationwide organ shortage and improving patient outcomes. While kidney transplants (KT) are performed more frequently, the overall number of patients on the waitlist consistently exceeds organ availability. Despite improved short-term outcomes in KT, comparable progress in long-term allograft survival has not been achieved. Major cause of graft loss at 5 years post-KT is chronic allograft dysfunction (CAD) characterized by interstitial fibrosis and tubular atrophy (IFTA). Accordingly, proactive prevention of CAD requires a comprehensive understanding of the immune mechanisms associated with either further dysfunction or impaired repair. Allograft rejection is primed by innate immune cells and carried out by adaptive immune cells. The rejection process is primarily facilitated by antibody-mediated rejection (ABMR) and T cell-mediated rejection (TCMR). It is essential to better elucidate the actions of individual immune cell subclasses (e.g. B memory, Tregs, Macrophage type 1 and 2) throughout the rejection process, rather than limiting our understanding to broad classes of immune cells. Embracing multi-omic approaches may be the solution in acknowledging these intricacies and decoding these enigmatic pathways. A transition alongside advancing technology will better allow organ biology to find its place in this era of precision and personalized medicine.
Collapse
Affiliation(s)
- Harsimar Kaur Ahuja
- Surgical Sciences Division, Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, U.S.A
| | - Shafquat Azim
- Surgical Sciences Division, Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, U.S.A
| | - Daniel Maluf
- Program of Transplantation, School of Medicine, 29S Greene St, University of Maryland, Baltimore, MD 21201, U.S.A
| | - Valeria R Mas
- Surgical Sciences Division, Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, U.S.A
| |
Collapse
|
7
|
Anwar IJ, Berman DM, DeLaura I, Gao Q, Willman MA, Miller A, Gill A, Gill C, Perrin S, Ricordi C, Ruiz P, Song M, Ladowski JM, Kirk AD, Kenyon NS. The anti-CD40L monoclonal antibody AT-1501 promotes islet and kidney allograft survival and function in nonhuman primates. Sci Transl Med 2023; 15:eadf6376. [PMID: 37647390 PMCID: PMC10990482 DOI: 10.1126/scitranslmed.adf6376] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 07/26/2023] [Indexed: 09/01/2023]
Abstract
Prior studies of anti-CD40 ligand (CD40L)-based immunosuppression demonstrated effective prevention of islet and kidney allograft rejection in nonhuman primate models; however, clinical development was halted because of thromboembolic complications. An anti-CD40L-specific monoclonal antibody, AT-1501 (Tegoprubart), was engineered to minimize risk of thromboembolic complications by reducing binding to Fcγ receptors expressed on platelets while preserving binding to CD40L. AT-1501 was tested in both a cynomolgus macaque model of intrahepatic islet allotransplantation and a rhesus macaque model of kidney allotransplantation. AT-1501 monotherapy led to long-term graft survival in both islet and kidney transplant models, confirming its immunosuppressive potential. Furthermore, AT-1501-based regimens after islet transplant resulted in higher C-peptide, greater appetite leading to weight gain, and reduced occurrence of cytomegalovirus reactivation compared with conventional immunosuppression. These data support AT-1501 as a safe and effective agent to promote both islet and kidney allograft survival and function in nonhuman primate models, warranting further testing in clinical trials.
Collapse
Affiliation(s)
- Imran J. Anwar
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | - Dora M. Berman
- Diabetes Research Institute, University of Miami; Miami, FL 33136, USA
- Department of Surgery, University of Miami; Miami, FL 33136, USA
| | - Isabel DeLaura
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | - Qimeng Gao
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | | | - Allison Miller
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | - Alan Gill
- ALS Therapy Development Institute; Cambridge, MA 02472, USA
| | - Cindy Gill
- ALS Therapy Development Institute; Cambridge, MA 02472, USA
| | | | - Camillo Ricordi
- Diabetes Research Institute, University of Miami; Miami, FL 33136, USA
- Department of Surgery, University of Miami; Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami; Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami; Miami, FL 33136, USA
- Department of Medicine, University of Miami; Miami, FL 33136, USA
| | - Philip Ruiz
- Department of Surgery, University of Miami; Miami, FL 33136, USA
| | - Mingqing Song
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | - Joseph M Ladowski
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | - Allan D. Kirk
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine; Durham, NC 27710, USA
| | - Norma S. Kenyon
- Diabetes Research Institute, University of Miami; Miami, FL 33136, USA
- Department of Surgery, University of Miami; Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami; Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami; Miami, FL 33136, USA
| |
Collapse
|
8
|
Miura S, Habibabady ZA, Pollok F, Ma M, Rosales IA, Kinoshita K, Pratts S, McGrath G, Chaban R, Fogarty S, Meibohm B, Daugherty B, Lederman S, Pierson RN. TNX-1500, a crystallizable fragment-modified anti-CD154 antibody, prolongs nonhuman primate cardiac allograft survival. Am J Transplant 2023; 23:1182-1193. [PMID: 37030662 PMCID: PMC10524282 DOI: 10.1016/j.ajt.2023.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023]
Abstract
Blockade of the CD40/CD154 T cell costimulation pathway is a promising approach to supplement or replace current clinical immunosuppression in solid organ transplantation. We evaluated the tolerability and activity of a novel humanized anti-CD154 monoclonal antibody, TNX-1500 (TNX), in a nonhuman primate heterotopic cardiac allogeneic (allo) transplant model. TNX-1500 contains a rupluzimab fragment antigen-binding region and an immunoglobin G4 crystallizable fragment region engineered to reduce binding to the crystallizable fragment gamma receptor IIa and associated risks of thrombosis. Recipients were treated for 6 months with standard-dose TNX (sTNX) monotherapy, low-dose TNX monotherapy (loTNX), or loTNX with mycophenolate mofetil (MMF) (loTNX + MMF). Results were compared with historical data using chimeric humanized 5c8 monotherapy dosed as for loTNX but discontinued at 3 months. Median survival time was similar for humanized 5c8 and both loTNX groups, but significantly longer with sTNX (>265 days) than with loTNX (99 days) or loTNX + MMF (88 days) (P < 0.05 for both comparisons against sTNX). Standard-dose TNX prevented antidonor alloantibody elaboration, inhibited chronic rejection, and was associated with a significantly reduced effector T cells/regulatory T cells ratio relative to loTNX with MMF. No thrombotic complications were observed. This study demonstrated that TNX was well tolerated, prolongs allograft survival, and prevents alloantibody production and cardiac allograft vasculopathy in a stringent preclinical nonhuman primate heart allotransplant model.
Collapse
Affiliation(s)
- Shuhei Miura
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Cardiovascular Surgery, Sapporo Medical University, Sapporo, Japan; Department of Cardiovascular Surgery, Teine Keijinkai Hospital, Sapporo, Japan.
| | - Zahra A Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Franziska Pollok
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Madelyn Ma
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shannon Pratts
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gannon McGrath
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan Chaban
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Bernd Meibohm
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | | - Richard N Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
9
|
Singh AK, Goerlich CE, Zhang T, Lewis BG, Hershfeld A, Mohiuddin MM. CD40-CD40L Blockade: Update on Novel Investigational Therapeutics for Transplantation. Transplantation 2023; 107:1472-1481. [PMID: 36584382 PMCID: PMC10287837 DOI: 10.1097/tp.0000000000004469] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Effective immune responses require antigen presentation by major histocompatibility complexes with cognate T-cell receptor and antigen-independent costimulatory signaling for T-cell activation, proliferation, and differentiation. Among several costimulatory signals, CD40-CD40L is of special interest to the transplantation community because it plays a vital role in controlling or regulating humoral and cellular immunity. Blockade of this pathway has demonstrated inhibition of donor-reactive T-cell responses and prolonged the survival of transplanted organs. Several anti-CD154 and anti-CD40 antibodies have been used in the transplantation model and demonstrated the potential of extending allograft and xenograft rejection-free survival. The wide use of anti-CD154 antibodies was hampered because of thromboembolic complications in transplant recipients. These antibodies have been modified to overcome the thromboembolic complications by altering the antibody binding fragment (Fab) and Fc (fragment, crystallizable) receptor region for therapeutic purposes. Here, we review recent preclinical advances to target the CD40-CD40L pair in transplantation.
Collapse
Affiliation(s)
| | | | - Tianshu Zhang
- University of Maryland School of Medicine, Baltimore, MD
| | | | | | | |
Collapse
|
10
|
Maenaka A, Kinoshita K, Hara H, Cooper DKC. The case for the therapeutic use of mechanistic/mammalian target of rapamycin (mTOR) inhibitors in xenotransplantation. Xenotransplantation 2023; 30:e12802. [PMID: 37029499 PMCID: PMC11286223 DOI: 10.1111/xen.12802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/23/2023] [Indexed: 04/09/2023]
Abstract
The mechanistic/mammalian target of rapamycin (mTOR) is one of the systems that are necessary to maintain cell homeostasis, such as survival, proliferation, and differentiation. mTOR inhibitors (mTOR-Is) are utilized as immunosuppressants and anti-cancer drugs. In organ allotransplantation, current regimens infrequently include an mTOR-I, which are positioned more commonly as alternative immunosuppressants. In clinical allotransplantation, long-term efficacy has been established, but there is a significant incidence of adverse events, for example, inhibition of wound healing, buccal ulceration, anemia, hyperglycemia, dyslipidemia, and thrombocytopenia, some of which are dose-dependent. mTOR-Is have properties that may be especially beneficial in xenotransplantation. These include suppression of T cell proliferation, increases in the number of T regulatory cells, inhibition of pig graft growth, and anti-inflammatory, anti-viral, and anti-cancer effects. We here review the potential benefits and risks of mTOR-Is in xenotransplantation and suggest that the benefits exceed the adverse effects.
Collapse
Affiliation(s)
- Akihiro Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Hidetaka Hara
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - David K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Reichart B, Cooper DKC, Längin M, Tönjes RR, Pierson RN, Wolf E. Cardiac xenotransplantation: from concept to clinic. Cardiovasc Res 2023; 118:3499-3516. [PMID: 36461918 PMCID: PMC9897693 DOI: 10.1093/cvr/cvac180] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 12/05/2022] Open
Abstract
For many patients with terminal/advanced cardiac failure, heart transplantation is the most effective, durable treatment option, and offers the best prospects for a high quality of life. The number of potentially life-saving donated human organs is far fewer than the population who could benefit from a new heart, resulting in increasing numbers of patients awaiting replacement of their failing heart, high waitlist mortality, and frequent reliance on interim mechanical support for many of those deemed among the best candidates but who are deteriorating as they wait. Currently, mechanical assist devices supporting left ventricular or biventricular heart function are the only alternative to heart transplant that is in clinical use. Unfortunately, the complication rate with mechanical assistance remains high despite advances in device design and patient selection and management, and the quality of life of the patients even with good outcomes is only moderately improved. Cardiac xenotransplantation from genetically multi-modified (GM) organ-source pigs is an emerging new option as demonstrated by the consistent long-term success of heterotopic (non-life-supporting) abdominal and life-supporting orthotopic porcine heart transplantation in baboons, and by a recent 'compassionate use' transplant of the heart from a GM pig with 10 modifications into a terminally ill patient who survived for 2 months. In this review, we discuss pig heart xenotransplantation as a concept, including pathobiological aspects related to immune rejection, coagulation dysregulation, and detrimental overgrowth of the heart, as well as GM strategies in pigs to prevent or minimize these problems. Additional topics discussed include relevant results of heterotopic and orthotopic heart transplantation experiments in the pig-to-baboon model, microbiological and virologic safety concepts, and efficacy requirements for initiating formal clinical trials. An adequate regulatory and ethical framework as well as stringent criteria for the selection of patients will be critical for the safe clinical development of cardiac xenotransplantation, which we expect will be clinically tested during the next few years.
Collapse
Affiliation(s)
- Bruno Reichart
- Walter Brendel Centre for Experimental Medicine, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - David K C Cooper
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Ralf R Tönjes
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, Langen 63225, Germany
| | - Richard N Pierson
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Eckhard Wolf
- Gene Centre and Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Munich 81377, Germany
| |
Collapse
|
12
|
Zhang W, Wang Y, Zhong F, Wang X, Sucher R, Lin CH, Brandacher G, Solari MG, Gorantla VS, Zheng XX. Donor derived hematopoietic stem cell niche transplantation facilitates mixed chimerism mediated donor specific tolerance. Front Immunol 2023; 14:1093302. [PMID: 36875068 PMCID: PMC9978155 DOI: 10.3389/fimmu.2023.1093302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
Compelling experimental evidence confirms that the robustness and longevity of mixed chimerism (MC) relies on the persistence and availability of donor-derived hematopoietic stem cell (HSC) niches in recipients. Based on our prior work in rodent vascularized composite allotransplantation (VCA) models, we hypothesize that the vascularized bone components in VCA bearing donor HSC niches, thus may provide a unique biologic opportunity to facilitate stable MC and transplant tolerance. In this study, by utilizing a series of rodent VCA models we demonstrated that donor HSC niches in the vascularized bone facilitate persistent multilineage hematopoietic chimerism in transplant recipients and promote donor-specific tolerance without harsh myeloablation. In addition, the transplanted donor HSC niches in VCA facilitated the donor HSC niches seeding to the recipient bone marrow compartment and contributed to the maintenance and homeostasis of stable MC. Moreover, this study provided evidences that chimeric thymus plays a role in MC-mediated transplant tolerance through a mechanism of thymic central deletion. Mechanistic insights from our study could lead to the use of vascularized donor bone with pre-engrafted HSC niches as a safe, complementary strategy to induce robust and stable MC-mediated tolerance in VCA or solid organ transplantation recipients.
Collapse
Affiliation(s)
- Wensheng Zhang
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yong Wang
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Fushun Zhong
- Transplantation Medical Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xinghuan Wang
- Transplantation Medical Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Robert Sucher
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mario G Solari
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Vijay S Gorantla
- Departments of Surgery, Ophthalmology and Bioengineering, Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Xin Xiao Zheng
- Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Transplantation Medical Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
13
|
Zhao J, Jung S, Li X, Li L, Kasinath V, Zhang H, Movahedi SN, Mardini A, Sabiu G, Hwang Y, Saxena V, Song Y, Ma B, Acton SE, Kim P, Madsen JC, Sage PT, Tullius SG, Tsokos GC, Bromberg JS, Abdi R. Delivery of costimulatory blockade to lymph nodes promotes transplant acceptance in mice. J Clin Invest 2022; 132:e159672. [PMID: 36519543 PMCID: PMC9754003 DOI: 10.1172/jci159672] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 10/11/2022] [Indexed: 12/15/2022] Open
Abstract
The lymph node (LN) is the primary site of alloimmunity activation and regulation during transplantation. Here, we investigated how fibroblastic reticular cells (FRCs) facilitate the tolerance induced by anti-CD40L in a murine model of heart transplantation. We found that both the absence of LNs and FRC depletion abrogated the effect of anti-CD40L in prolonging murine heart allograft survival. Depletion of FRCs impaired homing of T cells across the high endothelial venules (HEVs) and promoted formation of alloreactive T cells in the LNs in heart-transplanted mice treated with anti-CD40L. Single-cell RNA sequencing of the LNs showed that anti-CD40L promotes a Madcam1+ FRC subset. FRCs also promoted the formation of regulatory T cells (Tregs) in vitro. Nanoparticles (NPs) containing anti-CD40L were selectively delivered to the LNs by coating them with MECA-79, which binds to peripheral node addressin (PNAd) glycoproteins expressed exclusively by HEVs. Treatment with these MECA-79-anti-CD40L-NPs markedly delayed the onset of heart allograft rejection and increased the presence of Tregs. Finally, combined MECA-79-anti-CD40L-NPs and rapamycin treatment resulted in markedly longer allograft survival than soluble anti-CD40L and rapamycin. These data demonstrate that FRCs are critical to facilitating costimulatory blockade. LN-targeted nanodelivery of anti-CD40L could effectively promote heart allograft acceptance.
Collapse
Affiliation(s)
- Jing Zhao
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sungwook Jung
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaofei Li
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lushen Li
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vivek Kasinath
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hengcheng Zhang
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Said N. Movahedi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmad Mardini
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gianmarco Sabiu
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yoonha Hwang
- IVIM Technology, Daejeon, South Korea
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Vikas Saxena
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Bing Ma
- Institute for Genome Sciences and
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sophie E. Acton
- Stromal Immunology Group, Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Pilhan Kim
- IVIM Technology, Daejeon, South Korea
- Graduate School of Nanoscience and Technology and
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Joren C. Madsen
- Center for Transplantation Sciences, Department of Surgery
- Division of Cardiac Surgery, Department of Surgery, and
| | - Peter T. Sage
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - George C. Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Bromberg
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Reza Abdi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Li S, Xu H, Kirk AD. Modulation of Xenogeneic T-cell Proliferation by B7 and mTOR Blockade of T Cells and Porcine Endothelial Cells. Transplantation 2022; 106:950-962. [PMID: 34387242 PMCID: PMC8850983 DOI: 10.1097/tp.0000000000003920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Activation of porcine endothelial cells (PECs) is the mechanistic centerpiece of xenograft rejection. This study sought to characterize the immuno-phenotype of human T cells in response to PECs and to explore the immuno-modulation of B7 and mammalian target of rapamycin blockade of T cells and/or PECs during xeno-responses. METHODS Rapid memory T-cell (TM) responses to PECs were assessed by an intracellular cytokine staining. T-cell proliferation to PEC with or without belatacept or rapamycin was evaluated by a mixed lymphocyte-endothelial cell reaction (MLER). Additionally, rapamycin-pretreated PECs were used in MLER. Cell phenotypes were analyzed by flow cytometry. RESULTS Tumor necrosis factor-α/interferon-γ producers were detected in CD8+ cells stimulated by human endothelium but not PECs. MLER showed proliferation of CD4+ and CD8+ cells with predominantly memory subsets. Purified memory and naive cells proliferated following PEC stimulation with an increased frequency of TM in PEC-stimulated naive cells. Proliferating cells upregulated programmed cell death-1 (PD-1) and CD2 expression. Belatacept partially inhibited T-cell proliferation with reduced CD2 expression and frequency of the CD8+CD2highCD28- subset. Rapamycin dramatically inhibited PEC-induced T-cell proliferation, and rapamycin-preconditioned PECs failed to induce T-cell proliferation. PD-1 blockade did not restore T-cell proliferation to rapamycin-preconditioned PECs. CONCLUSIONS Humans lack rapid TM-mediated responses to PECs but induce T-cell proliferative responses characterized largely as TM with increasing CD2 and PD-1 expression. B7-CD28 and mammalian target of rapamycin blockade of T cells exhibit dramatic inhibitory effects in altering xeno-proliferating cells. Rapamycin alters PEC xeno-immunogenicity leading to inhibition of xeno-specific T-cell proliferation independent of PD-1-PD ligand interaction.
Collapse
Affiliation(s)
- Shu Li
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - He Xu
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Allan D. Kirk
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
15
|
Jin CY, Su N, Hu CB, Shao T, Ji JF, Qin LL, Fan DD, Lin AF, Xiang LX, Shao JZ. Regulatory role of BTLA and HVEM checkpoint inhibitors in T cell activation in a perciform fish Larimichthys crocea. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 128:104312. [PMID: 34767880 DOI: 10.1016/j.dci.2021.104312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 06/13/2023]
Abstract
The BTLA and HVEM are two well-characterized immune checkpoint inhibitors in humans and other mammalian species. However, the occurrence and functionality of these two molecules in non-mammalian species remain poorly understood. In the present study, we identified the BTLA and HVEM homologs from large yellow croaker (Larimichthys crocea), an economically important marine species of the perciform fish family. The Larimichthys crocea BTLA and HVEM (LcBTLA and LcHVEM) share conserved structural features to their mammalian counterparts, and they were expressed in various tissues and cells examined at different transcriptional levels, with particular abundance in immune-relevant tissues and splenic leukocytes. Immunofluorescence staining and flow cytometry analysis showed that LcHVEM and LcBTLA proteins were distributed on MHC-II+ APCs and CD4-2+ T cells, and a strong interaction between LcBTLA and LcHVEM was detected in splenic leukocytes in the mixed lymphocyte reaction (MLR). By blockade assays using anti-LcBTLA and anti-LcHVEM Abs as well as recombinant soluble LcBTLA and LcHVEM proteins in different combinations, it was found that LcBTLA-LcHVEM interactions play an important inhibitory role in the activation of alloreactive T cells using MLR as a model, and APC-initiated antigen-specific CD4-2+ T cells in response to A. hydrophila (A. h) stimulation. These observations highlight the extensive functional roles of LcBTLA and LcHVEM immune-checkpoint inhibitors in allogeneic T cell reactions, and CD4-2+ T cell-mediated adaptive immune responses in Larimichthys crocea. Thus, the BTLA-HVEM checkpoint may represent an ancient coinhibitory pathway, which was originated in fish and was conserved from fish to mammals throughout the vertebrate evolution.
Collapse
Affiliation(s)
- Chun-Yu Jin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Ning Su
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Chong-Bin Hu
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Tong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Jian-Fei Ji
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Lu-Lu Qin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Dong-Dong Fan
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Ai-Fu Lin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China
| | - Li-Xin Xiang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China.
| | - Jian-Zhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, People's Republic of China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, People's Republic of China.
| |
Collapse
|
16
|
Su N, Hu CB, Shao T, Jin CY, Li H, Ji JF, Qin LL, Fan DD, Lin AF, Xiang LX, Shao JZ. Functional role of CD40 and CD154 costimulatory signals in IgZ-mediated immunity against bacterial infection. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100038. [DOI: 10.1016/j.fsirep.2021.100038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 01/18/2023] Open
|
17
|
The posttraumatic response of CD4+ regulatory T cells is modulated by direct cell-cell contact via CD40L- and P-selectin-dependent pathways. Cent Eur J Immunol 2021; 46:283-294. [PMID: 34764800 PMCID: PMC8574106 DOI: 10.5114/ceji.2021.109171] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
CD4+ FoxP3+ regulatory T cells (CD4+ Tregs) are important for the posttraumatic anti-inflammatory host response. As described previously, platelets are able to modulate CD4+ Treg activity in a reciprocally activating interaction following injury. The underlying mechanisms of the posttraumatic interaction between platelets and CD4+ Tregs remain unclear. We investigated the potential influence of CD40L and P-selectin, molecules known to be involved in direct cell contact of these cell types. In a murine burn injury model, the potential interaction pathways were addressed using CD40L- and P-selectin-deficient mice. Draining lymph nodes were harvested following trauma (1 h) and following a sham procedure. Early rapid activation of CD4+ Tregs was assessed by phospho-flow cytometry (signaling molecules (p)PKC-δ and (p)ZAP-70). Platelet function was analyzed performing rotational thromboelastometry (ROTEM). We hypothesized that disruption of the direct cell-cell contact via CD40L and P-selectin would affect posttraumatic activation of CD4+ Tregs and influence the hemostatic function of platelets. Indeed, while injury induced early activation of CD4+ Tregs in wild-type mice (ZAP-70: p = 0.13, pZAP-70: p < 0.05, PKC-δ: p < 0.05, pPKC-δ: p < 0.05), disruption of CD40L-dependent interaction (ZAP-70: p = 0.57, pZAP-70: p = 0.68, PKC-δ: p = 0.68, pPKC-δ: p = 0.9) or P-selectin-dependent interaction (ZAP-70: p = 0.78, pZAP-70: p = 0.58, PKC-δ: p = 0.81, pPKC-δ: p = 0.73) resulted in reduced posttraumatic activation. Furthermore, hemostatic function was impaired towards hypocoagulability in either deficiency. Our results suggest that the posttraumatic activation of CD4+ Tregs and hemostatic function of platelets are affected by direct cell-cell-signaling via CD40L and P-selectin.
Collapse
|
18
|
Li W, Wang D, Yue R, Chen X, Liu A, Xu H, Teng P, Wang Z, Zou Y, Xu X, Zhao H, Li R, Fu Y, Guo L, Ni C, Fan J, Ma L. Gut microbes enlarged the protective effect of transplanted regulatory B cells on rejection of cardiac allografts. J Heart Lung Transplant 2021; 40:1502-1516. [PMID: 34742645 DOI: 10.1016/j.healun.2021.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 08/08/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Regulatory B cells (Bregs) play an important role in maintaining immune homeostasis and have the potential to induce tolerance. Previous work has found that Breg cells are involved in heart transplantation tolerance. However, the effect of Breg on the transplantation tolerance and the underlying mechanisms remain to be clarified. METHODS Using a within-species heart transplantation model, we aimed to investigate the role of CD19+CD5+CD1dhigh Bregs isolated from transplanted mice in preventing transplant rejection in vivo. We also explored the effects of CD40 and tumor necrosis factor receptor-associated factor 6 (TRAF6) ubiquitin ligase on Breg-mediated prolongation of survival in heart transplant (HT) mice, and the regulatory effects of downstream Cdk4 and Cdk6 proteins on dendritic cells (DCs), which clarified the function and molecular mechanism of Breg cells in HT mice. RESULTS Our data suggest that adoptive transfer of the transplanted Bregs served as an effective tolerance-inducing mechanism in HT mice and was involved in the CD40-TRAF6 signaling pathway in DCs. Moreover, DCs collected from the Breg treated HT mice also prolonged the survival of HT mice. Furthermore, DC-specific knockout of TRAF6 diminished Breg-mediated prolongation of survival in HT mice. Interestingly, gut microbes from donors increased the survival of cardiac allografts both in both the absence and presence of Bregs but were not implicated in CD40-TRAF6 signaling. CONCLUSIONS These findings reveal a role of Breg cells in the induction of transplantation tolerance through the blockade of the CD40-TRAF6 signaling pathway, which might be used in the treatment of HT in the clinic.
Collapse
Affiliation(s)
- Weidong Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dimin Wang
- School of Medicine, Zhejiang University, Hangzhou, China; Department of Reproductive endocrinology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Rongcai Yue
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Aixia Liu
- Department of Reproductive endocrinology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongfei Xu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Teng
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Zou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xingjie Xu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haige Zhao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Renyuan Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yufei Fu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lei Guo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengyao Ni
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingya Fan
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Ma
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
Reichart B, Längin M, Denner J, Schwinzer R, Cowan PJ, Wolf E. Pathways to Clinical Cardiac Xenotransplantation. Transplantation 2021; 105:1930-1943. [PMID: 33350675 DOI: 10.1097/tp.0000000000003588] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart transplantation is the only long-lasting lifesaving option for patients with terminal cardiac failure. The number of available human organs is however far below the actual need, resulting in substantial mortality of patients while waiting for a human heart. Mechanical assist devices are used to support cardiac function but are associated with a high risk of severe complications and poor quality of life for the patients. Consistent success in orthotopic transplantation of genetically modified pig hearts into baboons indicates that cardiac xenotransplantation may become a clinically applicable option for heart failure patients who cannot get a human heart transplant. In this overview, we project potential paths to clinical cardiac xenotransplantation, including the choice of genetically modified source pigs; associated requirements of microbiological, including virological, safety; optimized matching of source pig and recipient; and specific treatments of the donor heart after explantation and of the recipients. Moreover, selection of patients and the regulatory framework will be discussed.
Collapse
Affiliation(s)
- Bruno Reichart
- Walter Brendel Center for Experimental Medicine, LMU Munich, Munich, Germany
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Joachim Denner
- Institute of Virology, Free University Berlin, Berlin, Germany
| | - Reinhard Schwinzer
- Department of General-, Visceral-, and Transplantation Surgery, Transplant Laboratory, Hannover Medical School, Hannover, Germany
| | - Peter J Cowan
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, VIC, Australia
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
- Department of Veterinary Sciences, and Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| |
Collapse
|
20
|
Yanagawa S, Tahara H, Shirouzu T, Kawai S, Tanaka Y, Ide K, Akimoto S, Ohdan H. Development of a humanized mouse model to analyze antibodies specific for human leukocyte antigen (HLA). PLoS One 2021; 16:e0236614. [PMID: 33544740 PMCID: PMC7864411 DOI: 10.1371/journal.pone.0236614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 01/21/2021] [Indexed: 11/20/2022] Open
Abstract
In organ transplantation, human leukocyte antigen (HLA)-mismatch grafts not only induce the activation of cellular mediated immune response but also the development of chronic antibody-mediated rejection due to the donor-specific anti-HLA antibody (DSA) produced by B cells and plasma cells interacting with the graft endothelium. Significant improvement in long-term survival after transplantation can be expected if antibody-mediated rejection due to the DSA can be overcome. However, the mechanism of producing or controlling the DSA remains to be elucidated. In recent decades, “humanized” mouse models have been widely used for the basic research of human immune systems, but a humanized mouse model to analyze the mechanism of DSA production has not been established yet. Thus, we aimed to create a humanized mouse using a severe immunodeficiency mouse (NSG mouse) administered with human peripheral blood mononuclear cells (PBMCs). Initially, we detected a very low level of human total-IgG and no anti-HLA antibodies (Abs) in these mice. In our next attempt, we mixed PBMCs of various HLA antigenic combinations with or without regulatory T cells and preconditioned them by culturing on feeder cells stably transfected with human CD40 ligand (h-CD40L) alone or with h-CD40L and human B cell activating factor (h-BAFF). They were subsequently co-cultured with the corresponding irradiated stimulator PBMCs, and all cells were administered into naïve NSG mice. Although all three humanized models had sufficient human total-IgG and anti-HLA antibody production, allospecific anti-HLA Ab production was prominently suppressed whereas non-specific anti-HLA Abs were sufficiently detected. Therefore, this novel humanized mouse model might be useful for analyzing the mechanism of anti-allogeneic human B cell tolerance induction.
Collapse
Affiliation(s)
- Senichiro Yanagawa
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Tahara
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail:
| | - Takayuki Shirouzu
- Molecular Diagnostics Division, Wakunaga Pharmaceutical Co., Ltd., Osaka, Japan
| | - Shintaro Kawai
- Molecular Diagnostics Division, Wakunaga Pharmaceutical Co., Ltd., Osaka, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kentaro Ide
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuji Akimoto
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
21
|
CD40/CD40L Signaling as a Promising Therapeutic Target for the Treatment of Renal Disease. J Clin Med 2020; 9:jcm9113653. [PMID: 33202988 PMCID: PMC7697100 DOI: 10.3390/jcm9113653] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
The cluster of differentiation 40 (CD40) is activated by the CD40 ligand (CD40L) in a variety of diverse cells types and regulates important processes associated with kidney disease. The CD40/CD40L signaling cascade has been comprehensively studied for its roles in immune functions, whereas the signaling axis involved in local kidney injury has only drawn attention in recent years. Clinical studies have revealed that circulating levels of soluble CD40L (sCD40L) are associated with renal function in the setting of kidney disease. Levels of the circulating CD40 receptor (sCD40), sCD40L, and local CD40 expression are tightly related to renal injury in different types of kidney disease. Additionally, various kidney cell types have been identified as non-professional antigen-presenting cells (APCs) that express CD40 on the cell membrane, which contributes to the interactions between immune cells and local kidney cells during the development of kidney injury. Although the potential for adverse CD40 signaling in kidney cells has been reported in several studies, a summary of those studies focusing on the role of CD40 signaling in the development of kidney disease is lacking. In this review, we describe the outcomes of recent studies and summarize the potential therapeutic methods for kidney disease which target CD40.
Collapse
|
22
|
Reichart B, Längin M. On the way (my way) to clinical xenogeneic heart transplantation. Presented at the 15th biannual IXA meeting, Munich, October 11, 2019. Xenotransplantation 2020; 27:e12637. [DOI: 10.1111/xen.12637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023]
Affiliation(s)
- Bruno Reichart
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
| | - Matthias Längin
- Department of Anaesthesiology University Hospital LMU Munich Munich Germany
| |
Collapse
|
23
|
Zhao Y, Hu W, Chen P, Cao M, Zhang Y, Zeng C, Hara H, Cooper DKC, Mou L, Luan S, Gao H. Immunosuppressive and metabolic agents that influence allo‐ and xenograft survival by in vivo expansion of T regulatory cells. Xenotransplantation 2020; 27:e12640. [PMID: 32892428 DOI: 10.1111/xen.12640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/06/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Yanli Zhao
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center Institute of Translational Medicine Shenzhen University Health Science Center, Shenzhen University School of Medicine First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s Hospital Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | | | - Pengfei Chen
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Mengtao Cao
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Yingwei Zhang
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Changchun Zeng
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Hidetaka Hara
- Xenotransplantation Program Department of Surgery University of Alabama at Birmingham Birmingham AL USA
| | - David K. C. Cooper
- Xenotransplantation Program Department of Surgery University of Alabama at Birmingham Birmingham AL USA
| | - Lisha Mou
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center Institute of Translational Medicine Shenzhen University Health Science Center, Shenzhen University School of Medicine First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s Hospital Shenzhen China
| | - Shaodong Luan
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| | - Hanchao Gao
- Department of Nephrology Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
- Shenzhen Xenotransplantation Medical Engineering Research and Development Center Institute of Translational Medicine Shenzhen University Health Science Center, Shenzhen University School of Medicine First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s Hospital Shenzhen China
- Department of Medical Laboratory Shenzhen Longhua District Central Hospital Affiliated Central Hospital of Shenzhen Longhua District Guangdong Medical University Shenzhen China
| |
Collapse
|
24
|
Abstract
Costimulation between T cells and antigen-presenting cells is essential for the regulation of an effective alloimmune response and is not targeted with the conventional immunosuppressive therapy after kidney transplantation. Costimulation blockade therapy with biologicals allows precise targeting of the immune response but without non-immune adverse events. Multiple costimulation blockade approaches have been developed that inhibit the alloimmune response in kidney transplant recipients with varying degrees of success. Belatacept, an immunosuppressive drug that selectively targets the CD28-CD80/CD86 pathway, is the only costimulation blockade therapy that is currently approved for kidney transplant recipients. In the last decade, belatacept therapy has been shown to be a promising therapy in subgroups of kidney transplant recipients; however, the widespread use of belatacept has been tempered by an increased risk of acute kidney transplant rejection. The purpose of this review is to provide an overview of the costimulation blockade therapies that are currently in use or being developed for kidney transplant indications.
Collapse
|
25
|
Pig-to-non-human primate heart transplantation: The final step toward clinical xenotransplantation? J Heart Lung Transplant 2020; 39:751-757. [PMID: 32527674 DOI: 10.1016/j.healun.2020.05.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/25/2020] [Accepted: 05/10/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The demand for donated human hearts far exceeds the number available. Xenotransplantation of genetically modified porcine organs provides an alternative. In 2000, an Advisory Board of the International Society for Heart and Lung Transplantation set the benchmark for commencing clinical cardiac xenotransplantation as consistent 60% survival of non-human primates after life-supporting porcine heart transplantations. Recently, we reported the stepwise optimization of pig-to-baboon orthotopic cardiac xenotransplantation finally resulting in consistent success, with 4 recipients surviving 90 (n = 2), 182, and 195 days. Here, we report on 4 additional recipients, supporting the efficacy of our procedure. RESULTS The first 2 additional recipients succumbed to porcine cytomegalovirus (PCMV) infections on Days 15 and 27, respectively. In 2 further experiments, PCMV infections were successfully avoided, and 3-months survival was achieved. Throughout all the long-term experiments, heart, liver, and renal functions remained within normal ranges. Post-mortem cardiac diameters were slightly increased when compared with that at the time of transplantation but with no detrimental effect. There were no signs of thrombotic microangiopathy. The current regimen enabled the prolonged survival and function of orthotopic cardiac xenografts in altogether 6 of 8 baboons, of which 4 were now added. These results exceed the threshold set by the Advisory Board of the International Society for Heart and Lung Transplantation. CONCLUSIONS The results of our current and previous experimental cardiac xenotransplantations together fulfill for the first time the pre-clinical efficacy suggestions. PCMV-positive donor animals must be avoided.
Collapse
|
26
|
Schönberg A, Hamdorf M, Bock F. Immunomodulatory Strategies Targeting Dendritic Cells to Improve Corneal Graft Survival. J Clin Med 2020; 9:E1280. [PMID: 32354200 PMCID: PMC7287922 DOI: 10.3390/jcm9051280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/08/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
Even though the cornea is regarded as an immune-privileged tissue, transplantation always comes with the risk of rejection due to mismatches between donor and recipient. It is common sense that an alternative to corticosteroids as the current gold standard for treatment of corneal transplantation is needed. Since blood and lymphatic vessels have been identified as a severe risk factor for corneal allograft survival, much research has focused on vessel regression or inhibition of hem- and lymphangiogenesis in general. However, lymphatic vessels have been identified as required for the inflammation's resolution. Therefore, targeting other players of corneal engraftment could reveal new therapeutic strategies. The establishment of a tolerogenic microenvironment at the graft site would leave the recipient with the ability to manage pathogenic conditions independent from transplantation. Dendritic cells (DCs) as the central player of the immune system represent a target that allows the induction of tolerogenic mechanisms by many different strategies. These strategies are reviewed in this article with regard to their success in corneal transplantation.
Collapse
Affiliation(s)
- Alfrun Schönberg
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (A.S.); (M.H.)
| | - Matthias Hamdorf
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (A.S.); (M.H.)
| | - Felix Bock
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (A.S.); (M.H.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
27
|
Oh BC, Furtmüller GJ, Fryer ML, Guo Y, Messner F, Krapf J, Schneeberger S, Cooney DS, Lee WPA, Raimondi G, Brandacher G. Vascularized composite allotransplantation combined with costimulation blockade induces mixed chimerism and reveals intrinsic tolerogenic potential. JCI Insight 2020; 5:128560. [PMID: 32271163 DOI: 10.1172/jci.insight.128560] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
Vascularized composite allotransplantation (VCA) has become a valid therapeutic option to restore form and function after devastating tissue loss. However, the need for high-dose multidrug immunosuppression to maintain allograft survival is still hampering more widespread application of VCA. In this study, we investigated the immunoregulatory potential of costimulation blockade (CoB; CTLA4-Ig and anti-CD154 mAb) combined with nonmyeoablative total body irradiation (TBI) to promote allograft survival of VCA in a fully MHC-mismatched mouse model of orthotopic hind limb transplantation. Compared with untreated controls (median survival time [MST] 8 days) and CTLA4-Ig treatment alone (MST 17 days), CoB treatment increased graft survival (MST 82 days), and the addition of nonmyeloablative TBI led to indefinite graft survival (MST > 210 days). Our analysis suggests that VCA-derived BM induced mixed chimerism in animals treated with CoB and TBI + CoB, promoting gradual deletion of alloreactive T cells as the underlying mechanism of long-term allograft survival. Acceptance of donor-matched secondary skin grafts, decreased ex vivo T cell responsiveness, and increased graft-infiltrating Tregs further indicated donor-specific tolerance induced by TBI + CoB. In summary, our data suggest that vascularized BM-containing VCAs are immunologically favorable grafts promoting chimerism induction and long-term allograft survival in the context of CoB.
Collapse
Affiliation(s)
- Byoung Chol Oh
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Georg J Furtmüller
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Madeline L Fryer
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yinan Guo
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Hand and Microsurgery, Xiangya Hospital, Central South University, Hunan, China
| | - Franka Messner
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Visceral, Transplant and Thoracic Surgery, and
| | - Johanna Krapf
- Department of Plastic and Reconstructive Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Damon S Cooney
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - W P Andrew Lee
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Michaels AJ, Stoppato M, Flores WJ, Reimann KA, Engelman KD. Anti-CD40 antibody 2C10 binds to a conformational epitope at the CD40-CD154 interface that is conserved among primate species. Am J Transplant 2020; 20:298-305. [PMID: 31430418 PMCID: PMC6940519 DOI: 10.1111/ajt.15574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/28/2019] [Accepted: 08/09/2019] [Indexed: 01/25/2023]
Abstract
The antagonistic anti-CD40 antibody, 2C10, and its recombinant primate derivative, 2C10R4, are potent immunosuppressive antibodies whose utility in allo- and xenotransplantation have been demonstrated in nonhuman primate studies. In this study, we defined the 2C10 binding epitope and found only slight differences in affinity of 2C10 for CD40 derived from four primate species. Staining of truncation mutants mapped the 2C10 binding epitope to the N-terminal portion of CD40. Alanine scanning mutagenesis of the first 60 residues in the CD40 ectodomain highlighted key amino acids important for binding of 2C10 and for binding of the noncross-blocking anti-CD40 antibodies 3A8 and 5D12. All four 2C10-binding residues defined by mutagenesis clustered near the membrane-distal tip of CD40 and partially overlap the CD154 binding surface. In contrast, the overlapping 3A8 and 5D12 epitopes map to an opposing surface away from the CD154 binding domain. This biochemical characterization of 2C10 confirms the validity of nonhuman primate studies in the translation of this therapeutic antibody and provides insight its mechanism of action.
Collapse
Affiliation(s)
- Anthony J Michaels
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts
| | - Matteo Stoppato
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts
| | - Walter J Flores
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts
| | - Keith A Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts
| | - Kathleen D Engelman
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Yeung MY, Grimmig T, Sayegh MH. Costimulation Blockade in Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:267-312. [PMID: 31758538 DOI: 10.1007/978-981-32-9717-3_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
T cells play a pivotal role in orchestrating immune responses directed against a foreign (allogeneic) graft. For T cells to become fully activated, the T-cell receptor (TCR) must interact with the major histocompatibility complex (MHC) plus peptide complex on antigen-presenting cells (APCs), followed by a second "positive" costimulatory signal. In the absence of this second signal, T cells become anergic or undergo deletion. By blocking positive costimulatory signaling, T-cell allo-responses can be aborted, thus preventing graft rejection and promoting long-term allograft survival and possibly tolerance (Alegre ML, Najafian N, Curr Mol Med 6:843-857, 2006; Li XC, Rothstein DM, Sayegh MH, Immunol Rev 229:271-293, 2009). In addition, costimulatory molecules can provide negative "coinhibitory" signals that inhibit T-cell activation and terminate immune responses; strategies to promote these pathways can also lead to graft tolerance (Boenisch O, Sayegh MH, Najafian N, Curr Opin Organ Transplant 13:373-378, 2008). However, T-cell costimulation involves an incredibly complex array of interactions that may act simultaneously or at different times in the immune response and whose relative importance varies depending on the different T-cell subsets and activation status. In transplantation, the presence of foreign alloantigen incites not only destructive T effector cells but also protective regulatory T cells, the balance of which ultimately determines the fate of the allograft (Lechler RI, Garden OA, Turka LA, Nat Rev Immunol 3:147-158, 2003). Since the processes of alloantigen-specific rejection and regulation both require activation of T cells, costimulatory interactions may have opposing or synergistic roles depending on the cell being targeted. Such complexities present both challenges and opportunities in targeting T-cell costimulatory pathways for therapeutic purposes. In this chapter, we summarize our current knowledge of the various costimulatory pathways in transplantation and review the current state and challenges of harnessing these pathways to promote graft tolerance (summarized in Table 10.1).
Collapse
Affiliation(s)
- Melissa Y Yeung
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| | - Tanja Grimmig
- Department of Surgery, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Mohamed H Sayegh
- Department of Medicine, Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine and Immunology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
30
|
Mardomi A, Mohammadi N, Khosroshahi HT, Abediankenari S. An update on potentials and promises of T cell co-signaling molecules in transplantation. J Cell Physiol 2019; 235:4183-4197. [PMID: 31696513 DOI: 10.1002/jcp.29369] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
Abstract
The promising outcomes of immune-checkpoint based immunotherapies in cancer have provided a proportional perspective ahead of exploiting similar approaches in allotransplantation. Belatacept (CTLA-4-Ig) is an example of costimulation blockers successfully exploited in renal transplantation. Due to the wide range of regulatory molecules characterized in the past decades, some of these molecules might be candidates as immunomodulators in the case of tolerance induction in transplantation. Although there are numerous attempts on the apprehension of the effects of co-signaling molecules on immune response, the necessity for a better understanding is evident. By increasing the knowledge on the biology of co-signaling pathways, some pitfalls are recognized and improved approaches are proposed. The blockage of CD80/CD28 axis is an instance of evolution toward more efficacy. It is now evident that anti-CD28 antibodies are more effective than CD80 blockers in animal models of transplantation. Other co-signaling axes such as PD-1/PD-L1, CD40/CD154, 2B4/CD48, and others discussed in the present review are examples of critical immunomodulatory molecules in allogeneic transplantation. We review here the outcomes of recent experiences with co-signaling molecules in preclinical studies of solid organ transplantation.
Collapse
Affiliation(s)
- Alireza Mardomi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nabiallah Mohammadi
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Saeid Abediankenari
- Department of Immunology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
31
|
Flippe L, Bézie S, Anegon I, Guillonneau C. Future prospects for CD8 + regulatory T cells in immune tolerance. Immunol Rev 2019; 292:209-224. [PMID: 31593314 PMCID: PMC7027528 DOI: 10.1111/imr.12812] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD8+ Tregs have been long described and significant progresses have been made about their phenotype, their functional mechanisms, and their suppressive ability compared to conventional CD4+ Tregs. They are now at the dawn of their clinical use. In this review, we will summarize their phenotypic characteristics, their mechanisms of action, the similarities, differences and synergies between CD8+ and CD4+ Tregs, and we will discuss the biology, development and induction of CD8+ Tregs, their manufacturing for clinical use, considering open questions/uncertainties and future technically accessible improvements notably through genetic modifications.
Collapse
Affiliation(s)
- Léa Flippe
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
32
|
Zhang Y, Shen S, Zhao G, Xu CF, Zhang HB, Luo YL, Cao ZT, Shi J, Zhao ZB, Lian ZX, Wang J. In situ repurposing of dendritic cells with CRISPR/Cas9-based nanomedicine to induce transplant tolerance. Biomaterials 2019; 217:119302. [PMID: 31271858 DOI: 10.1016/j.biomaterials.2019.119302] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/27/2022]
Abstract
Organ transplantation is the only effective method to treat end-stage organ failure. However, it is continuously plagued by immune rejection, which is mostly caused by T cell-mediated reactions. Dendritic cells (DCs) are professional antigen-presenting cells, and blocking the costimulatory signaling molecule CD40 in DCs inhibits T cell activation and induces transplant tolerance. In this study, to relieve graft rejection, Cas9 mRNA (mCas9) and a guide RNA targeting the costimulatory molecule CD40 (gCD40) were prepared and encapsulated into poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PEG-b-PLGA)-based cationic lipid-assisted nanoparticles (CLAN), denoted CLANmCas9/gCD40. CLAN effectively delivered mCas9/gCD40 into DCs and disrupted CD40 in DCs at the genomic level both in vitro and in vivo. After intravenous injection into an acute mouse skin transplant model, CLANmCas9/gCD40-mediated CD40 disruption significantly inhibited T cell activation, which reduced graft damage and prolonged graft survival. This work provides a promising strategy for reprogramming DCs with nanoparticles carrying the CRISPR/Cas9 system to abate transplant rejection.
Collapse
Affiliation(s)
- Yue Zhang
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Song Shen
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Gui Zhao
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Cong-Fei Xu
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China.
| | - Hou-Bing Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, PR China
| | - Ying-Li Luo
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhi-Ting Cao
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China
| | - Jia Shi
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhi-Bin Zhao
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, 510006, PR China
| | - Jun Wang
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, PR China.
| |
Collapse
|
33
|
Schroder PM, Fitch ZW, Schmitz R, Choi AY, Kwun J, Knechtle SJ. The past, present, and future of costimulation blockade in organ transplantation. Curr Opin Organ Transplant 2019; 24:391-401. [PMID: 31157670 PMCID: PMC7088447 DOI: 10.1097/mot.0000000000000656] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Manipulating costimulatory signals has been shown to alter T cell responses and prolong graft survival in solid organ transplantation. Our understanding of and ability to target various costimulation pathways continues to evolve. RECENT FINDINGS Since the approval of belatacept in kidney transplantation, many additional biologics have been developed targeting clinically relevant costimulation signaling axes including CD40-CD40L, inducible costimulator-inducible costimulator ligand (ICOS-ICOSL), and OX40-OX40L. Currently, the effects of costimulation blockade on posttransplant humoral responses, tolerance induction, and xenotransplantation are under active investigation. Here, we will discuss these pathways as well as preclinical and clinical outcomes of biologics targeting these pathways in organ transplantation. SUMMARY Targeting costimultion is a promising approach for not only controlling T cell but also B cell responses. Consequently, costimulation blockade shows considerable potential for improving outcomes in antibody-mediated rejection and xenotransplantation.
Collapse
Affiliation(s)
- Paul M. Schroder
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Zachary W. Fitch
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Robin Schmitz
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Ashley Y. Choi
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA
| | | | | |
Collapse
|
34
|
DiChiacchio L, Singh AK, Chan JL, Shockcor NM, Zhang T, Lewis BG, Ayares D, Corcoran P, Horvath KA, Mohiuddin MM. Intra-Abdominal Heterotopic Cardiac Xenotransplantation: Pearls and Pitfalls. Front Cardiovasc Med 2019; 6:95. [PMID: 31404245 PMCID: PMC6669937 DOI: 10.3389/fcvm.2019.00095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/25/2019] [Indexed: 11/22/2022] Open
Abstract
Heterotopic cardiac xenotransplantation in the intra-abdominal position has been studied extensively in a pig-to-baboon model to define the optimal donor genetics and immunosuppressive regimen to prevent xenograft rejection. Extensive investigation using this model is a necessary stepping stone toward the development of a life-supporting animal model, with the ultimate goal of demonstrating suitability for clinical cardiac xenotransplantation trials. Aspects of surgical technique, pre- and post-operative care, graft monitoring, and minimization of infectious risk have all required refinement and optimization of heterotopic cardiac xenotransplantation over time. This review details non-immunologic obstacles relevant to this model described by our group and in the literature, as well as strategies that have been developed to address these specific challenges.
Collapse
Affiliation(s)
- Laura DiChiacchio
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Avneesh K. Singh
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Joshua L. Chan
- National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, United States
| | - Nicole M. Shockcor
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Tianshu Zhang
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Billeta G. Lewis
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | | | - Philip Corcoran
- National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, United States
| | - Keith A. Horvath
- National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, United States
| | - Muhammad M. Mohiuddin
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| |
Collapse
|
35
|
Fitch Z, Schmitz R, Kwun J, Hering B, Madsen J, Knechtle SJ. Transplant research in nonhuman primates to evaluate clinically relevant immune strategies in organ transplantation. Transplant Rev (Orlando) 2019; 33:115-129. [PMID: 31027947 PMCID: PMC6599548 DOI: 10.1016/j.trre.2019.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/08/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022]
Abstract
Research in transplant immunology using non-human primate (NHP) species to evaluate immunologic strategies to prevent rejection and prolong allograft survival has yielded results that have translated successfully into human organ transplant patient management. Other therapies have not proceeded to human translation due to failure in NHP testing, arguably sparing humans the futility and risk of such testing. The NHP transplant models are ethically necessary for drug development in this field and provide the closest analogue to human transplant patients available. The refinement of this resource with respect to colony MHC typing, reagent and assay development, and availability to the research community has greatly enhanced knowledge about transplant immunology and drug development.
Collapse
Affiliation(s)
- Zachary Fitch
- Department of Surgery, Duke Transplant Center, Durham, NC 27710, USA; Center for Transplantation Sciences, Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, White 510c, 55 Fruit Street, Boston, MA, USA
| | - Robin Schmitz
- Department of Surgery, Duke Transplant Center, Durham, NC 27710, USA
| | - Jean Kwun
- Department of Surgery, Duke Transplant Center, Durham, NC 27710, USA
| | - Bernhard Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Joren Madsen
- Department of Surgery, Duke Transplant Center, Durham, NC 27710, USA
| | - Stuart J Knechtle
- Department of Surgery, Duke Transplant Center, Durham, NC 27710, USA.
| |
Collapse
|
36
|
Selective CD28 Inhibition Modulates Alloimmunity and Cardiac Allograft Vasculopathy in Anti-CD154-Treated Monkeys. Transplantation 2018; 102:e90-e100. [PMID: 29319621 DOI: 10.1097/tp.0000000000002044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Selective CD28 inhibition is actively pursued as an alternative to B7 blockade using cytotoxic T lymphocyte antigen 4 Ig based on the hypothesis that the checkpoint immune regulators cytotoxic T lymphocyte antigen 4 and programmed death ligand 1 will induce tolerogenic immune signals. We previously showed that blocking CD28 using a monovalent nonactivating reagent (single-chain anti-CD28 Fv fragment linked to alpha-1 antitrypsin [sc28AT]) synergizes with calcineurin inhibitors in nonhuman primate (NHP) kidney and heart transplantation. Here, we explored the efficacy of combining a 3-week "induction" sc28AT treatment with prolonged CD154 blockade. METHODS Cynomolgus monkey heterotopic cardiac allograft recipients received sc28AT (10 mg/kg, d0-20, n = 3), hu5C8 (10-30 mg/kg, d0-84, n = 4), or combination (n = 6). Graft survival was monitored by telemetry. Protocol biopsies and graft explants were analyzed for International Society of Heart and Lung Transplantation acute rejection grade and cardiac allograft vasculopathy score. Alloantibody, T-cell phenotype and regulatory T cells were analyzed by flow cytometry. Immunochemistry and gene expression (NanoString) characterized intra-graft cellular infiltration. RESULTS Relative to modest prolongation of median graft survival time with sc28AT alone (34 days), hu5C8 (133 days), and sc28AT + hu5C8 (141 days) prolonged survival to a similar extent. CD28 blockade at induction, added to hu5C8, significantly attenuated the severity of acute rejection and cardiac allograft vasculopathy during the first 3 months after transplantation relative to hu5C8 alone. These findings were associated with decreased proportions of circulating CD8 and CD3CD28 T cells, and modulation of inflammatory gene expression within allografts. CONCLUSIONS Induction with sc28AT promotes early cardiac allograft protection in hu5C8-treated NHPs. These results support further investigation of prolonged selective CD28 inhibition with CD40/CD154 blockade in NHP transplants.
Collapse
|
37
|
Lorente L. New prognostic biomarkers of mortality in patients undergoing liver transplantation for hepatocellular carcinoma. World J Gastroenterol 2018; 24:4230-4242. [PMID: 30310256 PMCID: PMC6175764 DOI: 10.3748/wjg.v24.i37.4230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/18/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
The outcome prediction of hepatocellular carcinoma (HCC) patients undergoing liver transplantation (LT) was classically established using various macromorphological factors and serum alpha-fetoprotein levels prior to LT. However, other biomarkers have recently been reported to be associated with the prognosis of HCC patients undergoing to LT. This review summarizes clinical data on these new biomarkers. High blood levels of malondialdehyde, total antioxidant capacity, caspase-cleaved cytokeratin-18, soluble CD40 ligand, substance P, C-reactive protein, and vascular endothelial growth factor, increased neutrophil to lymphocyte ratio and platelet to lymphocyte ratio in blood, high peripheral blood expression of human telomerase reverse transcriptase messenger ribonucleic acid, and high HCC expression of dickkopf-1 have recently been associated with decreased survival rates. In addition, high blood levels of des-gamma-carboxy prothrombin, and high HCC expression of glypican-3, E-cadherin and beta-catenin have been associated with increased HCC recurrence. Additional research is necessary to establish the prognostic role of these biomarkers in HCC prior to LT. Furthermore, some of these biomarkers are also interesting because their potential modulation could help to create new research lines for improving the outcomes of those patients.
Collapse
Affiliation(s)
- Leonardo Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, Santa Cruz de Tenerife 38320, Spain
| |
Collapse
|
38
|
Fähnrich A, Klein S, Sergé A, Nyhoegen C, Kombrink S, Möller S, Keller K, Westermann J, Kalies K. CD154 Costimulation Shifts the Local T-Cell Receptor Repertoire Not Only During Thymic Selection but Also During Peripheral T-Dependent Humoral Immune Responses. Front Immunol 2018; 9:1019. [PMID: 29867987 PMCID: PMC5966529 DOI: 10.3389/fimmu.2018.01019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/24/2018] [Indexed: 12/20/2022] Open
Abstract
CD154 is a transmembrane cytokine expressed transiently on activated CD4 T cells upon T-cell receptor (TCR) stimulation that interacts with CD40 on antigen-presenting cells. The signaling via CD154:CD40 is essential for B-cell maturation and germinal center formation and also for the final differentiation of CD4 T cells during T-dependent humoral immune responses. Recent data demonstrate that CD154 is critically involved in the selection of T-cell clones during the negative selection process in the thymus. Whether CD154 signaling influences the TCR repertoire during peripheral T-dependent humoral immune responses has not yet been elucidated. To find out, we used CD154-deficient mice and assessed the global TCRβ repertoire in T-cell zones (TCZ) of spleens by high-throughput sequencing after induction of a Th2 response to the multiepitopic antigen sheep red blood cells. Qualitative and quantitative comparison of the splenic TCZ-specific TCRβ repertoires revealed that CD154 deficiency shifts the distribution of Vβ-Jβ genes after antigen exposure. This data led to the conclusion that costimulation via CD154:CD40 during the interaction of T cells with CD40-matured B cells contributes to the recruitment of T-cell clones into the immune response and thereby shapes the peripheral TCR repertoire.
Collapse
Affiliation(s)
- Anke Fähnrich
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Sebastian Klein
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Arnauld Sergé
- Centre de Recherche en Cancérologie de Marseille (CRCM) U1068 INSERM - UMR7258 CNRS - Institut Paoli Calmette, Aix-Marseille University, UM105, Marseille, France
| | | | - Sabrina Kombrink
- Institute of Mathematics, University of Luebeck, Luebeck, Germany
| | - Steffen Möller
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Karsten Keller
- Institute of Mathematics, University of Luebeck, Luebeck, Germany
| | | | - Kathrin Kalies
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| |
Collapse
|
39
|
Ock SA, Oh KB, Hwang S, Yun IJ, Ahn C, Chee HK, Kim H, Ullah I, Im GS, Park EW. Immune molecular profiling of whole blood drawn from a non-human primate cardiac xenograft model treated with anti-CD154 monoclonal antibodies. Xenotransplantation 2018; 25:e12392. [PMID: 29582477 DOI: 10.1111/xen.12392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 01/17/2018] [Accepted: 02/22/2018] [Indexed: 12/17/2022]
Abstract
Most studies of xenografts have been carried out with complex immunosuppressive regimens to prevent immune rejection; however, such treatments may be fatal owing to unknown causes. Here, we performed immune molecular profiling following anti-CD154 monoclonal antibody (mAb) treatment in heterotopic abdominal cardiac xenografts from α-1,3-galactosyltransferase-knockout pigs into cynomolgus monkeys to elucidate the mechanisms mediating the undesirable fatal side effects of immunosuppressive agents. Blood samples were collected from healthy monkeys as control and then at 2 days after xenograft transplantation and just before humane euthanasia; 94 genes related to the immune system were analyzed. The basic immunosuppressive regimen included cobra venom factor, anti-thymocyte globulin, and rituximab, with and without anti-CD154 mAbs. The maintenance therapy was followed with tacrolimus, MMF, and methylprednisolone. The number of upregulated genes was initially decreased on Day 2 (-/+ anti-CD154 mAb, 22/13) and then increased before euthanasia in recipients treated with anti-CD154 mAbs (-/+ anti-CD154 mAb, 30/37). The number of downregulated genes was not affected by anti-CD154 mAb treatment. Additionally, the number of upregulated genes increased over time for both groups. Interestingly, treatment with anti-CD154 mAbs upregulated coagulation inducers (CCL2/IL6) before euthanasia. In conclusion, immunosuppressive regimens used for cardiac xenografting affected upregulation of 6 inflammation genes (CXCL10, MPO, MYD88, NLRP3, TNFα, and TLR1) and downregulation of 8 genes (CCR4, CCR6, CD40, CXCR3, FOXP3, GATA3, STAT4, and TBX21).
Collapse
Affiliation(s)
- Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Seongsoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Ik Jin Yun
- Department of Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Curie Ahn
- Division of Nephrology, Seoul National University College of Medicine, Seoul, Korea.,Designed Animal & Transplantation Research institute, Institute of Green BioScience & Technology, Seoul National University, Pyeongchang, Gangwon-do, Korea
| | - Hyun Ken Chee
- Department of Cardiothoracic Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Hwajung Kim
- Division of Nephrology, Seoul National University College of Medicine, Seoul, Korea
| | - Imran Ullah
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Gi-Sun Im
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Eung Woo Park
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| |
Collapse
|
40
|
Rachakonda SP, Dai H, Penack O, Blau O, Blau IW, Radujkovic A, Müller-Tidow C, Kumar R, Dreger P, Luft T. Single Nucleotide Polymorphisms in CD40L Predict Endothelial Complications and Mortality After Allogeneic Stem-Cell Transplantation. J Clin Oncol 2018; 36:789-800. [DOI: 10.1200/jco.2017.76.4662] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Purpose Endothelial vulnerability is a potential risk factor for complications after allogeneic stem-cell transplantation (alloSCT). The CD40/CD40 ligand (CD40L) axis contributes to inflammatory diseases and is upregulated in endothelial cells upon activation, suggesting a role in alloSCT biology. Here, we studied single nucleotide polymorphisms (SNPs) in the CD40L gene in recipients of alloSCT. Patients and Methods Three CD40L SNPs (rs3092920, rs3092952, rs3092936) were analyzed for association with transplant-associated thrombotic microangiopathy, overall nonrelapse mortality (NRM), and NRM after acute graft-versus-host disease in 294 recipients of alloSCT without statin-based endothelial prophylaxis (SEP). The significant genotype was then put into perspective with established thrombomodulin ( THBD) gene polymorphisms. Findings were validated in an independent cohort without SEP and in an additional 344 patients who received SEP. Results The rs3092936 CC/CT genotype was associated with an increased risk of transplant-associated thrombotic microangiopathy ( P = .001), overall NRM ( P = .03), and NRM after acute graft-versus-host disease ( P = .01). The rs3092936 CC/CT genotype was largely mutually exclusive of high-risk THBD SNPs. Both CD40L and THBD SNPs predicted adverse overall survival (OS) and overall NRM to a similar extent in training cohort (OS, P = .04; NRM, P < .001) and validation cohort (OS, P = .01; NRM, P = .001) without SEP. In contrast, SEP completely abolished the influence of the high-risk CD40L and THBD SNPs ( P = .40). Conclusion An increased risk of endothelial complications can be predicted before alloSCT by genetic markers in the recipient’s genome. The normalization of mortality risks in patients treated with SEP suggests a way of overcoming the negative effect of high-risk genotypes and warrants further clinical validation.
Collapse
Affiliation(s)
- Sivaramakrishna P. Rachakonda
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Hao Dai
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Olaf Penack
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Olga Blau
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Igor Wolfgang Blau
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Aleksandar Radujkovic
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Carsten Müller-Tidow
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Rajiv Kumar
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Peter Dreger
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| | - Thomas Luft
- Sivaramakrishna P. Rachakonda, Hao Dai, and Rajiv Kumar, German Cancer Research Centre; Sivaramakrishna P. Rachakonda, Aleksandar Radujkovic, Carsten Müller-Tidow, Peter Dreger, and Thomas Luft, University Hospital Heidelberg, Heidelberg; and Olaf Penack, Olga Blau, and Igor Wolfgang Blau, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
41
|
Li T, Shi Y, Sun W, Wang H, Wang Q, Jiang Y. Increased PD-1 +CD154 + Tfh cells are possibly the most important functional subset of PD-1 + T follicular helper cells in adult patients with minimal change disease. Mol Immunol 2017; 94:98-106. [PMID: 29288900 DOI: 10.1016/j.molimm.2017.12.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/18/2017] [Accepted: 12/22/2017] [Indexed: 12/11/2022]
Abstract
T follicular helper (Tfh) cells, especially programmed cell death protein 1 (PD-1)+ Tfh cells, exert important functions in the normal immune response. The purpose of this study was to determine the frequency of different subsets of PD-1+ Tfh cells and their functional effects in adult patients with minimal change disease (MCD). The frequencies of circulating PD-1+, PD-1+CD154+, and PD-1+interleukin (IL)-21+ Tfh cells, and CD38+CD19+ and CD38+CD19+CD40+ B cells, as well as serum IL-2, IL-4, IL-17A, IL-6, IL-21, and interferon (IFN)-γ were significantly increased in the MCD patients compared with the healthy controls (HCs) (P < 0.05). However, no significant difference was found in PD-1+BCL-6+ or PD-1+ICOS+ Tfh cells. Furthermore, the percentages of PD-1+ Tfh and PD-1+CD154+ Tfh cells were negatively correlated with the estimated glomerular filtration rate (eGFR), but positively correlated with the 24-h urinary protein concentration and serum IL-21 level. The percentages of PD-1+ Tfh and PD-1+CD154+ Tfh cells were positively correlated with the percentages of CD38+ plasma cells and active CD38+CD40+ plasma cells, respectively. After an 8-12-week treatment with prednisolone, the percentages of PD-1+, PD-1+CD154+, and PD-1+IL-21+ Tfh cells as well as the serum level of IL-21 were significantly reduced; in contrast, the serum levels of IL-4 and IL-10 were increased (P < 0.05). We conclude that increased PD-1+CD154+ Tfh cells are possibly the most important functional subset of PD-1+ Tfh cells and may contribute towards the pathogenesis of MCD.
Collapse
Affiliation(s)
- Tao Li
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yunpeng Shi
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Haifeng Wang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Quan Wang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China; Key Laboratory of Zoonoses Research, Ministry of Education, The First Hospital of Jilin University, Changchun, 130021, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
42
|
Pilat N, Granofszky N, Wekerle T. Combining Adoptive Treg Transfer with Bone Marrow Transplantation for Transplantation Tolerance. CURRENT TRANSPLANTATION REPORTS 2017; 4:253-261. [PMID: 29201599 PMCID: PMC5691126 DOI: 10.1007/s40472-017-0164-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The mixed chimerism approach is an exceptionally potent strategy for the induction of donor-specific tolerance in organ transplantation and so far the only one that was demonstrated to work in the clinical setting. Regulatory T cells (Tregs) have been shown to improve chimerism induction in experimental animal models. This review summarizes the development of innovative BMT protocols using therapeutic Treg transfer for tolerance induction. RECENT FINDINGS Treg cell therapy promotes BM engraftment in reduced conditioning protocols in both, mice and non-human primates. In mice, transfer of polyclonal recipient Tregs was sufficient to substitute cytotoxic recipient conditioning. Treg therapy prevented chronic rejection of skin and heart allografts related to tissue-specific antigen disparities, in part by promoting intragraft Treg accumulation. SUMMARY Adoptive Treg transfer is remarkably effective in facilitating BM engraftment in reduced-intensity protocols in mice and non-human primates. Furthermore, it promotes regulatory mechanisms that prevent chronic rejection.
Collapse
Affiliation(s)
- Nina Pilat
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Nicolas Granofszky
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Thomas Wekerle
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
43
|
High serum soluble CD40L levels previously to liver transplantation in patients with hepatocellular carcinoma are associated with mortality at one year. J Crit Care 2017; 43:316-320. [PMID: 29020665 DOI: 10.1016/j.jcrc.2017.09.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/29/2017] [Accepted: 09/15/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE CD40L and its soluble form (sCD40L) are proteins of the tumor necrosis factor superfamily (TNFSF) that exhibit prothrombotic and proinflammatory properties when binding to CD40, which is a cell surface receptor of the tumor necrosis factor receptor superfamily (TNFRSF). High circulating levels of sCD40L have been associated with poor prognosis in patients with hepatocellular carcinoma (HCC). However, it is unknown whether there is an association between circulating sCD40L levels and survival in patients with HCC underwent to liver transplantation (LT), and this was the objective of that study. METHODS Serum sCD40L levels were measured in a total of 139 patients before LT (124 survivors at 1year of LT and 15 non-survivors). The end-point study was 1year survival after liver LT. RESULTS We found that 1-year non-surviving patients showed higher serum sCD40L levels than survivor patients (p=0.02). We found in logistic regression analysis that serum sCD40L levels higher than 321pg/mL (Odds Ratio=6.86; 95% confidence interval=2.06-22.76; p=0.002) and age of LT deceased donor were associated with death at 1year. CONCLUSIONS The new finding of our study was that high serum sCD40L levels previously to LT in patients with HCC are associated with higher mortality at one year.
Collapse
|
44
|
O'Neill NA, Zhang T, Braileanu G, Sun W, Cheng X, Hershfeld A, Laird CT, Kronfli A, Hock LA, Dahi S, Kubicki N, Sievert E, Hassanein W, Cimeno A, Pierson RN, Azimzadeh AM. Comparative Evaluation of αCD40 (2C10R4) and αCD154 (5C8H1 and IDEC-131) in a Nonhuman Primate Cardiac Allotransplant Model. Transplantation 2017; 101:2038-2047. [PMID: 28557955 PMCID: PMC5568940 DOI: 10.1097/tp.0000000000001836] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Specific blockade of T cell costimulation pathway is a promising immunomodulatory approach being developed to replace our current clinical immunosuppression therapies. The goal of this study is to compare results associated with 3 monoclonal antibodies directed against the CD40/CD154 T cell costimulation pathway. METHODS Cynomolgus monkey heterotopic cardiac allograft recipients were treated with either IDEC-131 (humanized αCD154, n = 9), 5C8H1 (mouse-human chimeric αCD154, n = 5), or 2C10R4 (mouse-rhesus chimeric αCD40, n = 6) monotherapy using a consistent, comparable dosing regimen for 3 months after transplant. RESULTS Relative to the previously reported IDEC-131-treated allografts, median survival time (35 ± 31 days) was significantly prolonged in both 5C8H1-treated (142 ± 26, P < 0.002) and 2C10R4-treated (124 ± 37, P < 0.020) allografts. IDEC-131-treated grafts had higher cardiac allograft vasculopathy severity scores during treatment relative to either 5C8H1 (P = 0.008) or 2C10R4 (P = 0.0002). Both 5C8H1 (5 of 5 animals, P = 0.02) and 2C10R4 (6/6, P = 0.007), but not IDEC-131 (2/9), completely attenuated IgM antidonor alloantibody (alloAb) production during treatment; 5C8H1 (5/5) more consistently attenuated IgG alloAb production compared to 2C10R4 (4/6) and IDEC-131 (0/9). All evaluable explanted grafts experienced antibody-mediated rejection. Only 2C10R4-treated animals exhibited a modest, transient drop in CD20 lymphocytes from baseline at day 14 after transplant (-457 ± 152 cells/μL) compared with 5C8H1-treated animals (16 ± 25, P = 0.037), and the resurgent B cells were primarily of a naive phenotype. CONCLUSIONS In this model, CD154/CD40 axis blockade using IDEC-131 is an inferior immunomodulatory treatment than 5C8H1 or 2C10R4, which have similar efficacy to prolong graft survival and to delay cardiac allograft vasculopathy development and antidonor alloAb production during treatment.
Collapse
Affiliation(s)
- Natalie A. O'Neill
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Tianshu Zhang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Gheorghe Braileanu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Wenji Sun
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Xiangfei Cheng
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Alena Hershfeld
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | | | - Anthony Kronfli
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Lindsay A. Hock
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - Sia Dahi
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Natalia Kubicki
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Evelyn Sievert
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Wessam Hassanein
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Arielle Cimeno
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Richard N. Pierson
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Agnes M. Azimzadeh
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
45
|
LEA29Y expression in transgenic neonatal porcine islet-like cluster promotes long-lasting xenograft survival in humanized mice without immunosuppressive therapy. Sci Rep 2017; 7:3572. [PMID: 28620237 PMCID: PMC5472587 DOI: 10.1038/s41598-017-03913-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/05/2017] [Indexed: 02/02/2023] Open
Abstract
Genetically engineered pigs are a promising source for islet cell transplantation in type 1 diabetes, but the strong human anti-pig immune response prevents its successful clinical application. Here we studied the efficacy of neonatal porcine islet-like cell clusters (NPICCs) overexpressing LEA29Y, a high-affinity variant of the T cell co-stimulation inhibitor CTLA-4Ig, to engraft and restore normoglycemia after transplantation into streptozotocin-diabetic NOD-SCID IL2rγ−/− (NSG) mice stably reconstituted with a human immune system. Transplantation of INSLEA29Y expressing NPICCs resulted in development of normal glucose tolerance (70.4%) and long-term maintenance of normoglycemia without administration of immunosuppressive drugs. All animals transplanted with wild-type NPICCs remained diabetic. Immunohistological examinations revealed a strong peri- and intragraft infiltration of wild-type NPICCs with human CD45+ immune cells consisting of predominantly CD4+ and CD8+ lymphocytes and some CD68+ macrophages and FoxP3+ regulatory T cells. Significantly less infiltrating lymphocytes and only few macrophages were observed in animals transplanted with INSLEA29Y transgenic NPICCs. This is the first study providing evidence that beta cell-specific LEA29Y expression is effective for NPICC engraftment in the presence of a humanized immune system and it has a long-lasting protective effect on inhibition of human anti-pig xenoimmunity. Our findings may have important implications for the development of a low-toxic protocol for porcine islet transplantation in patients with type 1 diabetes.
Collapse
|
46
|
Preemptive CD20+ B cell Depletion Attenuates Cardiac Allograft Vasculopathy in CD154-Treated Monkeys. Transplantation 2016; 101:63-73. [PMID: 27362307 DOI: 10.1097/tp.0000000000001258] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Anti-CD154 monotherapy is associated with antidonor allo-antibody (Ab) elaboration, cardiac allograft vasculopathy (CAV), and allograft failure in preclinical primate cell and organ transplant models. In the context of calcineurin inhibitors (CNI), these pathogenic phenomena are delayed by preemptive "induction" B cell depletion. METHODS αCD154 (IDEC-131)-treated cynomolgus monkey heart allograft recipients were given peritransplant rituximab (αCD20) alone or with rabbit antihuman thymocyte globulin. RESULTS Relative to previously reported reference groups, αCD20 significantly prolonged survival, delayed Ab detection, and attenuated CAV within 3 months in αCD154-treated recipients (αCD154 + αCD20 graft median survival time > 90 days, n = 7, vs 28 days for αCD154 alone (IDEC-131), n = 21; P = 0.05). Addition of rabbit antihuman thymocyte globulin to αCD154 (n = 6) or αCD154 + αCD20 (n = 10) improved graft protection from graft rejection and failure during treatment but was associated with significant morbidity in 8 of 16 recipients (6 infections, 2 drug-related complications). In αCD20-treated animals, detection of antidonor Ab and relatively severe CAV were anticipated by appearance of CD20 cells (>1% of lymphocytes) in peripheral blood and were associated with low αCD154 trough levels (below 100 μg/mL). CONCLUSIONS These observations support the hypothesis that efficient preemptive "induction" CD20 B cell depletion consistently modulates pathogenic alloimmunity and attenuates CAV in this translational model, extending our prior findings with calcineurin inhibitors to the context of CD154 blockade.
Collapse
|