1
|
Alegre ML, Atkinson C, Issa F, Valujskikh A, Zhang ZJ. BEST PRACTICES OF HEART TRANSPLANTATION IN MICE. Am J Transplant 2025:S1600-6135(25)00217-5. [PMID: 40252924 DOI: 10.1016/j.ajt.2025.04.012] [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: 01/09/2025] [Revised: 03/21/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Heart transplantation in mice has served as a reliable in vivo model in transplant research worldwide for more than half a century. It is not only useful for addressing cardiac graft-specific questions but also provides mechanistic insights and therapeutic strategies that have broad impact across all solid organ transplants. Compared to other mouse models of solid organ transplantation, such as kidney, lung, or small intestine transplants, the surgical techniques to perform mouse heart transplantation (mHT) are relatively easy to master, and the graft heartbeat offers a simple means to evaluate transplant viability. However, as with other in vivo mouse models, mHT has distinct strengths and limitations. Multiple factors can influence the accuracy and reproducibility of the results, including microsurgical techniques and microsurgeons' skills, post-op monitoring methodologies, mouse strain combinations, sex/age. As innovative biotechnologies continue to emerge, the future holds many opportunities for preclinical research utilizing the mHT model. It is therefore imperative to provide the field with optimized mHT protocols and maintain standard reporting requirements. This minireview provides a concise summary and recommendations for standardized practices to ensure the accuracy, reproducibility, and translational value of findings generated from mHT model.
Collapse
Affiliation(s)
- Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL, USA
| | - Carl Atkinson
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH; Cleveland Clinic Lerner College of Medicine, Cleveland, OH, United States
| | - Zheng Jenny Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
2
|
Kawai T, Williams WW, Elias N, Fishman JA, Crisalli K, Longchamp A, Rosales IA, Duggan M, Kimura S, Morena L, Borges TJ, Tomosugi T, Karadagi A, Nakamura T, Safa K, Giarraputo A, Avillach CT, Patalas ED, Smith RN, Sachs DH, Cosimi AB, Madsen JC, Cooper DKC, Pierson R, Perrin S, Anand RP, Chhangawala S, Coscarella M, Daigneault A, Li F, Pearce O, Qin W, Serkin WT, Yeung V, Getchell K, Low SC, Curtis M, Colvin RB, Riella LV. Xenotransplantation of a Porcine Kidney for End-Stage Kidney Disease. N Engl J Med 2025. [PMID: 39927618 DOI: 10.1056/nejmoa2412747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Xenotransplantation offers a potential solution to the organ shortage crisis. A 62-year-old hemodialysis-dependent man with long-standing diabetes, advanced vasculopathy, and marked dialysis-access challenges received a gene-edited porcine kidney with 69 genomic edits, including deletion of three glycan antigens, inactivation of porcine endogenous retroviruses, and insertion of seven human transgenes. The xenograft functioned immediately. The patient's creatinine levels decreased promptly and progressively, and dialysis was no longer needed. After a T-cell-mediated rejection episode on day 8, intensified immunosuppression reversed rejection. Despite sustained kidney function, the patient died from unexpected, sudden cardiac causes on day 52; autopsy revealed severe coronary artery disease and ventricular scarring without evident xenograft rejection. (Funded by Massachusetts General Hospital and eGenesis.).
Collapse
Affiliation(s)
- Tatsuo Kawai
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Winfred W Williams
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Nahel Elias
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Jay A Fishman
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Kerry Crisalli
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Alban Longchamp
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Michael Duggan
- Center for Comparative Medicine, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Shoko Kimura
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Leela Morena
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Thiago J Borges
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Toshihide Tomosugi
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Ahmad Karadagi
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Tsukasa Nakamura
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Kassem Safa
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Alessia Giarraputo
- Department of Pathology, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Claire T Avillach
- Department of Pathology, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Eva D Patalas
- Department of Pathology, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - R Neal Smith
- Department of Pathology, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - David H Sachs
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - A Benedict Cosimi
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Joren C Madsen
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - David K C Cooper
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Richard Pierson
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Robert B Colvin
- Department of Pathology, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| | - Leonardo V Riella
- Transplant Center, Massachusetts General Hospital, Boston
- Harvard Medical School, Boston
| |
Collapse
|
3
|
Sanatkar SA, Kinoshita K, Maenaka A, Hara H, Cooper DKC. The Evolution of Immunosuppressive Therapy in Pig-to-Nonhuman Primate Organ Transplantation. Transpl Int 2025; 37:13942. [PMID: 39872238 PMCID: PMC11770881 DOI: 10.3389/ti.2024.13942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/27/2024] [Indexed: 01/30/2025]
Abstract
An overview is provided of the evolution of strategies towards xenotransplantation during the past almost 40 years, focusing on advances in gene-editing of the organ-source pigs, pre-transplant treatment of the recipient, immunosuppressive protocols, and adjunctive therapy. Despite initial challenges, including hyperacute rejection resulting from natural (preformed) antibody binding and complement activation, significant progress has been made through gene editing of the organ-source pigs and refinement of immunosuppressive regimens. Major steps were the identification and deletion of expression of the three known glycan xenoantigens on pig vascular endothelial cells, the transgenic expression of human "protective" proteins, e.g., complement-regulatory, coagulation-regulatory, and anti-inflammatory proteins, and the administration of an immunosuppressive regimen based on blockade of the CD40/CD154 T cell co-stimulation pathway. Efforts to address systemic inflammation followed. The synergy between gene editing and judicious immunomodulation appears to largely prevent graft rejection and is associated with a relatively good safety profile. Though there remains an incidence of severe or persistent proteinuria (nephrotic syndrome) in a minority of cases. This progress offers renewed hope for patients in need of life-saving organ transplants.
Collapse
Affiliation(s)
- S. A. Sanatkar
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - K. Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - A. Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - H. Hara
- The Transplantation Institute at the Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - D. K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Marwedel B, Medina LY, De May H, Adogla JE, Kennedy E, Flores E, Lim E, Adams S, Bartee E, Serda RE. Regional immune mechanisms enhance efficacy of an autologous cellular cancer vaccine with intraperitoneal administration. Oncoimmunology 2024; 13:2421029. [PMID: 39625271 PMCID: PMC11540083 DOI: 10.1080/2162402x.2024.2421029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 12/06/2024] Open
Abstract
Widespread peritoneal dissemination is common in patients with gynecologic or gastrointestinal cancers. Accumulating evidence of a central role for regional immunity in cancer control indicates that intraperitoneal immunotherapy may have treatment advantages. This study delineates immune mechanisms engaged by intraperitoneal delivery of a cell-based vaccine comprised of silicified ovarian cancer cells associated with enhanced survival. Vaccine trafficking from the site of injection to milky spots and other fat-associated lymphoid clusters was studied in syngeneic cancer models using bioluminescent and fluorescent imaging, microscopy, and flow cytometry. Spectral flow cytometry was used to phenotype peritoneal immune cell populations, while bioluminescent imaging of cancer was used to study myeloid and T cell dependency, systemic immunity, and vaccine efficacy in models of disseminated high-grade serous ovarian and DNA mismatch-repair proficient microsatellite-stable colorectal cancer. Following intraperitoneal vaccination of mice with ovarian cancer, vaccine cells were rapidly internalized by myeloid cells, with subsequent trafficking to fat-associated lymphoid clusters. Tumor clearance was confirmed to be T cell-mediated, leading to the establishment of local and systemic immunity. Combination immune checkpoint inhibitor and vaccine therapy in mice with advanced disease, characterized by an established suppressive tumor microenvironment, increased the number of mice with non-detectable tumors, however, change in tumor burden compared to vaccine monotherapy was not significant. Vaccination also resulted in tumor clearance in mouse models of metastatic colorectal cancer. This study demonstrates that intraperitoneal vaccine delivery has the potential to enhance vaccine efficacy by activating resident immune cells with the subsequent establishment of protective systemic anti-tumor immunity.
Collapse
Affiliation(s)
- Ben Marwedel
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Lorél Y. Medina
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Henning De May
- Department of Obstetrics & Gynecology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Joshua E. Adogla
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Ellie Kennedy
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Erica Flores
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Eunju Lim
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Sarah Adams
- Department of Obstetrics & Gynecology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Eric Bartee
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Rita E. Serda
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| |
Collapse
|
5
|
Bender M, Abicht JM, Reichart B, Neumann E, Radan J, Mokelke M, Buttgereit I, Leuschen M, Wall F, Michel S, Ellgass R, Steen S, Paskevicius A, Lange A, Kessler B, Kemter E, Klymiuk N, Denner J, Godehardt AW, Tönjes RR, Burgmann JM, Figueiredo C, Milusev A, Zollet V, Salimi-Afjani N, Despont A, Rieben R, Ledderose S, Walz C, Hagl C, Ayares D, Wolf E, Schmoeckel M, Brenner P, Binder U, Gebauer M, Skerra A, Längin M. Combination of Anti-CD40 and Anti-CD40L Antibodies as Co-Stimulation Blockade in Preclinical Cardiac Xenotransplantation. Biomedicines 2024; 12:1927. [PMID: 39200391 PMCID: PMC11351779 DOI: 10.3390/biomedicines12081927] [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: 06/20/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The blockade of the CD40/CD40L immune checkpoint is considered essential for cardiac xenotransplantation. However, it is still unclear which single antibody directed against CD40 or CD40L (CD154), or which combination of antibodies, is better at preventing organ rejection. For example, the high doses of antibody administered in previous experiments might not be feasible for the treatment of humans, while thrombotic side effects were described for first-generation anti-CD40L antibodies. To address these issues, we conducted six orthotopic pig-to-baboon cardiac xenotransplantation experiments, combining a chimeric anti-CD40 antibody with an investigational long-acting PASylated anti-CD40L Fab fragment. The combination therapy effectively resulted in animal survival with a rate comparable to a previous study that utilized anti-CD40 monotherapy. Importantly, no incidence of thromboembolic events associated with the administration of the anti-CD40L PAS-Fab was observed. Two experiments failed early because of technical reasons, two were terminated deliberately after 90 days with the baboons in excellent condition and two were extended to 120 and 170 days, respectively. Unexpectedly, and despite the absence of any clinical signs, histopathology revealed fungal infections in all four recipients. This study provides, for the first time, insights into a combination therapy with anti-CD40/anti-CD40L antibodies to block this immune checkpoint.
Collapse
Affiliation(s)
- Martin Bender
- Department of Anaesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Jan-Michael Abicht
- Department of Anaesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Bruno Reichart
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, 81377 Munich, Germany
| | - Elisabeth Neumann
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, 81377 Munich, Germany
| | - Julia Radan
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, 81377 Munich, Germany
| | - Maren Mokelke
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, 81377 Munich, Germany
| | - Ines Buttgereit
- Department of Anaesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Maria Leuschen
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, 81377 Munich, Germany
| | - Felicia Wall
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, 81377 Munich, Germany
| | - Sebastian Michel
- Department of Cardiac Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Heart Alliance, German Center for Cardiovascular Research (DZHK), 81377 Munich, Germany
| | - Reinhard Ellgass
- Department of Cardiac Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Stig Steen
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, 22242 Lund, Sweden
| | - Audrius Paskevicius
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, 22242 Lund, Sweden
| | - Andreas Lange
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
| | - Barbara Kessler
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
| | - Elisabeth Kemter
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
| | - Joachim Denner
- Institute of Virology, Free University Berlin, 14163 Berlin, Germany
| | - Antonia W. Godehardt
- Division of Haematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Ralf R. Tönjes
- Division of Haematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Jonathan M. Burgmann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Constança Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, 30625 Hannover, Germany
| | - Anastasia Milusev
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3008 Bern, Switzerland
| | - Valentina Zollet
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3008 Bern, Switzerland
| | - Neda Salimi-Afjani
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3008 Bern, Switzerland
| | - Alain Despont
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Stephan Ledderose
- Institute of Pathology, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Christoph Walz
- Institute of Pathology, Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Christian Hagl
- Department of Cardiac Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Heart Alliance, German Center for Cardiovascular Research (DZHK), 81377 Munich, Germany
| | | | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 81377 Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany
| | - Michael Schmoeckel
- Department of Cardiac Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Paolo Brenner
- Department of Cardiac Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Uli Binder
- XL-protein GmbH, 85354 Freising, Germany
| | | | - Arne Skerra
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany
| |
Collapse
|
6
|
Kwun J, Kirk AD, Knechtle SJ. The emerging era of organ transplantation and anti-CD154mAb. Am J Transplant 2024; 24:1339-1341. [PMID: 38643943 DOI: 10.1016/j.ajt.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Affiliation(s)
- Jean Kwun
- Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA.
| | - Allan D Kirk
- Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Stuart J Knechtle
- Duke Transplant Center, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
7
|
Liu D, Yao H, Ferrer IR, Ford ML. Differential induction of donor-reactive Foxp3 + regulatory T cell via blockade of CD154 vs CD40. Am J Transplant 2024; 24:1369-1381. [PMID: 38552961 PMCID: PMC11305915 DOI: 10.1016/j.ajt.2024.03.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/30/2024]
Abstract
Recently published studies in both murine models and a meta-analysis of non-human primate renal transplant studies showed that anti-CD154 reagents conferred a significant survival advantage over CD40 blockers in both animal models and across multiple organs. Here we sought to compare the induction of donor-reactive forkhead box P3+-induced regulatory T cells (Foxp3+ iTreg) in mice treated with anti-CD154 versus anti-CD40 monoclonal antibodies (mAbs). Results indicated that while treatment with anti-CD154 mAb resulted in a significant increase in the frequency of donor-reactive CD4+ Foxp3+ iTreg following transplantation, treatment with anti-CD40 or Cd40 deficiency failed to recapitulate this result. Because we recently identified CD11b as an alternate receptor for CD154 during alloimmunity, we interrogated the role of CD154:CD11b interactions in the generation of Foxp3+ iTreg and found that blockade of CD11b in Cd40-/- recipients resulted in increased donor-reactive Foxp3+ iTreg as compared with CD40 deficiency alone. Mechanistically, CD154:CD11b inhibition decreased interleukin (IL)-1β from CD11b+ and CD11c+ dendritic cells, and blockade of IL-1β synergized with CD40 deficiency to promote Foxp3+ iTreg induction and prolong allograft survival. Taken together, these data provide a mechanistic basis for the observed inferiority of anti-CD40 blockers as compared with anti-CD154 mAb and illuminate an IL-1β-dependent mechanism by which CD154:CD11b interactions prevent the generation of donor-reactive Foxp3+ iTreg during transplantation.
Collapse
Affiliation(s)
- Danya Liu
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, Georgia, USA
| | - Hongmin Yao
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, Georgia, USA
| | - Ivana R Ferrer
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, Georgia, USA
| | - Mandy L Ford
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
8
|
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
|
9
|
Yue H, Bai L. Progress, implications, and challenges in using humanized immune system mice in CAR-T therapy-Application evaluation and improvement. Animal Model Exp Med 2024; 7:3-11. [PMID: 37823214 PMCID: PMC10961865 DOI: 10.1002/ame2.12353] [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: 06/15/2023] [Accepted: 09/17/2023] [Indexed: 10/13/2023] Open
Abstract
In recent years, humanized immune system (HIS) mice have been gradually used as models for preclinical research in pharmacotherapies and cell therapies with major breakthroughs in tumor and other fields, better mimicking the human immune system and the tumor immune microenvironment, compared to traditional immunodeficient mice. To better promote the application of HIS mice in preclinical research, we selectively summarize the current prevalent and breakthrough research and evaluation of chimeric antigen receptor (CAR) -T cells in various antiviral and antitumor treatments. By exploring its application in preclinical research, we find that it can better reflect the actual clinical patient condition, with the advantages of providing high-efficiency detection indicators, even for progressive research and development. We believe that it has better clinical patient simulation and promotion for the updated design of CAR-T cell therapy than directly transplanted immunodeficient mice. The characteristics of the main models are proposed to improve the use defects of the existing models by reducing the limitation of antihost reaction, combining multiple models, and unifying sources and organoid substitution. Strategy study of relapse and toxicity after CAR-T treatment also provides more possibilities for application and development.
Collapse
Affiliation(s)
- Hanwei Yue
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal SciencesCAMS and PUMCChao‐yang District, BeijingChina
| | - Lin Bai
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal SciencesCAMS and PUMCChao‐yang District, BeijingChina
| |
Collapse
|
10
|
Abstract
Despite significant advances in the field of transplantation in the past two decades, current clinically available therapeutic options for immunomodulation remain fairly limited. The advent of calcineurin inhibitor-based immunosuppression has led to significant success in improving short-term graft survival; however, improvements in long-term graft survival have stalled. Solid organ transplantation provides a unique opportunity for immunomodulation of both the donor organ prior to implantation and the recipient post transplantation. Furthermore, therapies beyond targeting the adaptive immune system have the potential to ameliorate ischemic injury to the allograft and halt its aging process, augment its repair, and promote recipient immune tolerance. Other recent advances include expanding the donor pool by reducing organ discard, and bioengineering and genetically modifying organs from other species to generate transplantable organs. Therapies discussed here will likely be most impactful if individualized on the basis of specific donor and recipient considerations.
Collapse
Affiliation(s)
- Irma Husain
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA;
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA;
- Duke Transplant Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
11
|
Abstract
Memory T cells that are specific for alloantigen can arise from a variety of stimuli, ranging from direct allogeneic sensitization from prior transplantation, blood transfusion, or pregnancy to the elicitation of pathogen-specific T cells that are cross-reactive with alloantigen. Regardless of the mechanism by which they arise, alloreactive memory T cells possess key metabolic, phenotypic, and functional properties that render them distinct from naive T cells. These properties affect the immune response to transplantation in 2 important ways: first, they can alter the speed, location, and effector mechanisms with which alloreactive T cells mediate allograft rejection, and second, they can alter T-cell susceptibility to immunosuppression. In this review, we discuss recent developments in understanding these properties of memory T cells and their implications for transplantation.
Collapse
Affiliation(s)
| | - Mandy L. Ford
- Emory Transplant Center, Emory University, Atlanta, GA
| |
Collapse
|
12
|
Zeng S, Crichton ES, Ford ML, Badell IR. Memory T follicular helper cells drive donor-specific antibodies independent of memory B cells and primary germinal center and alloantibody formation. Am J Transplant 2023; 23:1511-1525. [PMID: 37302575 PMCID: PMC11228286 DOI: 10.1016/j.ajt.2023.06.006] [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] [Received: 04/14/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Human leukocyte antigen antibodies are important immunologic mediators of renal allograft loss and are difficult to control. The inability to permanently eliminate donor-specific antibodies (DSA) is partly due to an incomplete understanding of the cellular mechanisms driving alloantibody formation, recurrence, and maintenance. Memory T follicular helper (mTfh) cells rapidly interact with memory B cells upon antigen re-exposure for anamnestic humoral responses, but little is known about Tfh memory in transplantation. We hypothesized that alloreactive mTfh cells form after transplantation and play a critical role in DSA formation following alloantigen re-encounter. To test this hypothesis, we utilized murine skin allograft models to identify and characterize Tfh memory and interrogate its ability to mediate alloantibody responses. We identified alloreactive Tfh memory as a mediator of accelerated humoral alloresponses independent of memory B cells and primary germinal center, or DSA, formation. Furthermore, we demonstrate that mTfh-driven alloantibody formation is susceptible to CD28 costimulation blockade. These findings provide novel insight into a pathologic role for memory Tfh in alloantibody responses and strongly support shifting therapeutic focus from the singular targeting of B cell lineage cells and alloantibodies themselves to multimodal strategies that include inhibition of mTfh cells to treat DSA.
Collapse
Affiliation(s)
- Shan Zeng
- Emory Transplant Center, Atlanta, Georgia, USA
| | | | | | | |
Collapse
|
13
|
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: 2.5] [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
|
14
|
Lassiter G, Otsuka R, Hirose T, Rosales I, Karadagi A, Tomosugi T, Dehnadi A, Lee H, Colvin RB, Baardsnes J, Moraitis A, Smith EE, Ali Z, Berhe P, Mulder A, Meibohm B, Daugherty B, Fogarty S, Pierson RN, Lederman S, Kawai T. TNX-1500, a crystallizable fragment-modified anti-CD154 antibody, prolongs nonhuman primate renal allograft survival. Am J Transplant 2023; 23:1171-1181. [PMID: 37019335 PMCID: PMC10527606 DOI: 10.1016/j.ajt.2023.03.022] [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: 02/08/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023]
Abstract
The blockade of the CD154-CD40 pathway with anti-CD154 monoclonal antibody has been a promising immunomodulatory approach to prevent allograft rejection. However, clinical trials of immunoglobulin G1 antibodies targeting this pathway revealed thrombogenic properties, which were subsequently shown to be mediated by crystallizable fragment (Fc)-gamma receptor IIa-dependent platelet activation. To prevent thromboembolic complications, an immunoglobulin G4 anti-CD154 monoclonal antibody, TNX-1500, which retains the fragment antigen binding region of ruplizumab (humanized 5c8, BG9588), was modified by protein engineering to decrease Fc binding to Fc-gamma receptor IIa while retaining certain other effector functions and pharmacokinetics comparable with natural antibodies. Here, we report that TNX-1500 treatment is not associated with platelet activation in vitro and consistently inhibits kidney allograft rejection in vivo without clinical or histologic evidence of prothrombotic phenomena. We conclude that TNX-1500 retains efficacy similar to that of 5c8 to prevent kidney allograft rejection while avoiding previously identified pathway-associated thromboembolic complications.
Collapse
Affiliation(s)
- Grace Lassiter
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryo Otsuka
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Takayuki Hirose
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Rosales
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmad Karadagi
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Toshihide Tomosugi
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abbas Dehnadi
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Lee
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert B Colvin
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Anna Moraitis
- National Research Council, Montréal, Quebec H4P 2R2, Canada
| | - Emma E Smith
- National Research Council, Montréal, Quebec H4P 2R2, Canada
| | - Zahida Ali
- Charles River Laboratories, Skokie, Illinois, USA
| | - Phil Berhe
- Charles River Laboratories, Skokie, Illinois, USA
| | | | - Bernd Meibohm
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennesse, USA
| | | | | | - Richard N Pierson
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Seth Lederman
- Tonix Pharmaceuticals, Inc, Chatham, New Jersey, USA
| | - Tatsuo Kawai
- Center for Transplantation Sciences, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
15
|
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: 0.5] [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
|
16
|
Unger LW, Muckenhuber M, Mahr B, Schwarz C, Pilat N, Granofszky N, Regele H, Wekerle T. Chronic CD40L blockade is required for long-term cardiac allograft survival with a clinically relevant CTLA4-Ig dosing regimen. Front Immunol 2022; 13:1060576. [PMID: 36569922 PMCID: PMC9773869 DOI: 10.3389/fimmu.2022.1060576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction In de-novo kidney transplantation, the CTLA4-Ig fusion protein belatacept is associated with improved graft function but also an increased risk of acute rejection compared to calcineurin inhibitor therapy. The combination with a second costimulation blocker could potentially improve outcome while avoiding calcineurin inhibitor toxicity. The aim of this study was to define the conditions under which the combination of CTLA4-Ig and CD40L blockade leads to rejection-free permanent graft survival in a stringent murine heart transplantation model. Methods Naïve wild-type or CD40L (CD154) knock-out mice received a fully mismatched BALB/c cardiac allograft. Selected induction and maintenance protocols for CTLA4-Ig and blocking αCD40L monoclonal antibodies (mAB) were investigated. Graft survival, rejection severity and donor-specific antibody (DSA) formation were assessed during a 100-day follow-up period. Results and Discussion Administering αCD40L mAb as monotherapy at the time of transplantation significantly prolonged heart allograft survival but did not further improve the outcome when given in addition to chronic CTLA4-Ig therapy (which prolongs graft survival to a median of 22 days). Likewise, chronic αCD40L mAb therapy (0.5mg) combined with perioperative CTLA4-Ig led to rejection in a proportion of mice and extensive histological damage, despite abrogating DSA formation. Only the permanent interruption of CD40-CD40L signaling by using CD40L-/- recipient mice or by chronic αCD40L administration synergized with chronic CTLA4-Ig to achieve long-term allograft survival with preserved histological graft integrity in all recipients without DSA formation. The combination of α-CD40L and CTLA4-Ig works most effectively when both therapeutics are administered chronically.
Collapse
Affiliation(s)
- Lukas W. Unger
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Moritz Muckenhuber
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Benedikt Mahr
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Christoph Schwarz
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Nina Pilat
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Nicolas Granofszky
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| | - Heinz Regele
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Department of General Surgery, Division of Transplantation, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Duneton C, Winterberg PD, Ford ML. Activation and regulation of alloreactive T cell immunity in solid organ transplantation. Nat Rev Nephrol 2022; 18:663-676. [PMID: 35902775 PMCID: PMC9968399 DOI: 10.1038/s41581-022-00600-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 01/18/2023]
Abstract
Transplantation is the only curative treatment for patients with kidney failure but it poses unique immunological challenges that must be overcome to prevent allograft rejection and ensure long-term graft survival. Alloreactive T cells are important contributors to graft rejection, and a clearer understanding of the mechanisms by which these cells recognize donor antigens - through direct, indirect or semi-direct pathways - will facilitate their therapeutic targeting. Post-T cell priming rejection responses can also be modified by targeting pathways that regulate T cell trafficking, survival cytokines or innate immune activation. Moreover, the quantity and quality of donor-reactive memory T cells crucially shape alloimmune responses. Of note, many fundamental concepts in transplant immunology have been derived from models of infection. However, the programmed differentiation of allograft-specific T cell responses is probably distinct from that of pathogen-elicited responses, owing to the dearth of pathogen-derived innate immune activation in the transplantation setting. Understanding the fundamental (and potentially unique) immunological pathways that lead to allograft rejection is therefore a prerequisite for the rational development of therapeutics that promote transplantation tolerance.
Collapse
Affiliation(s)
- Charlotte Duneton
- Paediatric Nephrology, Robert Debré Hospital, Paris, France
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Pamela D Winterberg
- Paediatric Nephrology, Emory University Department of Paediatrics and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mandy L Ford
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
18
|
Iglesias M, Brennan DC, Larsen CP, Raimondi G. Targeting inflammation and immune activation to improve CTLA4-Ig-based modulation of transplant rejection. Front Immunol 2022; 13:926648. [PMID: 36119093 PMCID: PMC9478663 DOI: 10.3389/fimmu.2022.926648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
For the last few decades, Calcineurin inhibitors (CNI)-based therapy has been the pillar of immunosuppression for prevention of organ transplant rejection. However, despite exerting effective control of acute rejection in the first year post-transplant, prolonged CNI use is associated with significant side effects and is not well suited for long term allograft survival. The implementation of Costimulation Blockade (CoB) therapies, based on the interruption of T cell costimulatory signals as strategy to control allo-responses, has proven potential for better management of transplant recipients compared to CNI-based therapies. The use of the biologic cytotoxic T-lymphocyte associated protein 4 (CTLA4)-Ig is the most successful approach to date in this arena. Following evaluation of the BENEFIT trials, Belatacept, a high-affinity version of CTLA4-Ig, has been FDA approved for use in kidney transplant recipients. Despite its benefits, the use of CTLA4-Ig as a monotherapy has proved to be insufficient to induce long-term allograft acceptance in several settings. Multiple studies have demonstrated that events that induce an acute inflammatory response with the consequent release of proinflammatory cytokines, and an abundance of allograft-reactive memory cells in the recipient, can prevent the induction of or break established immunomodulation induced with CoB regimens. This review highlights advances in our understanding of the factors and mechanisms that limit CoB regimens efficacy. We also discuss recent successes in experimentally designing complementary therapies that favor CTLA4-Ig effect, affording a better control of transplant rejection and supporting their clinical applicability.
Collapse
Affiliation(s)
- Marcos Iglesias
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| | - Daniel C. Brennan
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Christian P. Larsen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Giorgio Raimondi
- Vascularized and Composite Allotransplantation (VCA) Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Giorgio Raimondi, ; Marcos Iglesias,
| |
Collapse
|
19
|
Muckenhuber M, Wekerle T, Schwarz C. Costimulation blockade and Tregs in solid organ transplantation. Front Immunol 2022; 13:969633. [PMID: 36119115 PMCID: PMC9478950 DOI: 10.3389/fimmu.2022.969633] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
Regulatory T cells (Tregs) play a critical role in maintaining self-tolerance and in containing allo-immune responses in the context of transplantation. Recent advances yielded the approval of the first pharmaceutical costimulation blockers (abatacept and belatacept), with more of them in the pipeline. These costimulation blockers inhibit effector cells with high clinical efficacy to control disease activity, but might inadvertently also affect Tregs. Treg homeostasis is controlled by a complex network of costimulatory and coinhibitory signals, including CD28, the main target of abatacept/belatacept, and CTLA4, PD-1 and ICOS. This review shall give an overview on what effects the therapeutic manipulation of costimulation has on Treg function in transplantation.
Collapse
Affiliation(s)
- Moritz Muckenhuber
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
- *Correspondence: Thomas Wekerle, ; Christoph Schwarz,
| | - Christoph Schwarz
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
- *Correspondence: Thomas Wekerle, ; Christoph Schwarz,
| |
Collapse
|
20
|
Novel Functions of Integrins as Receptors of CD154: Their Role in Inflammation and Apoptosis. Cells 2022; 11:cells11111747. [PMID: 35681441 PMCID: PMC9179867 DOI: 10.3390/cells11111747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022] Open
Abstract
CD154, an inflammatory mediator also known as CD40 ligand, has been identified as a novel binding partner for some members of the integrin family. The αIIbβ3, specifically expressed on platelets, was the first integrin to be described as a receptor for CD154 after CD40. Its interaction with soluble CD154 (sCD154) highly contributes to thrombus formation and stability. Identifying αIIbβ3 opened the door for investigating other integrins as partners of CD154. The αMβ2 expressed on myeloid cells was shown capable of binding CD154 and contributing as such to cell activation, adhesion, and release of proinflammatory mediators. In parallel, α5β1 communicates with sCD154, inducing pro-inflammatory responses. Additional pathogenic effects involving apoptosis-preventing functions were exhibited by the CD154–α5β1 dyad in T cells, conferring a role for such interaction in the survival of malignant cells, as well as the persistence of autoreactive T cells. More recently, CD154 receptors integrated two new integrin members, αvβ3 and α4β1, with little known as to their biological significance in this context. This article provides an overview of the novel role of integrins as receptors of CD154 and as critical players in pro-inflammatory and apoptotic responses.
Collapse
|
21
|
Perrin S, Magill M. The Inhibition of CD40/CD154 Costimulatory Signaling in the Prevention of Renal Transplant Rejection in Nonhuman Primates: A Systematic Review and Meta Analysis. Front Immunol 2022; 13:861471. [PMID: 35464470 PMCID: PMC9022482 DOI: 10.3389/fimmu.2022.861471] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022] Open
Abstract
The prevention of allograft transplant rejection by inhibition of the CD40/CD40L costimulatory pathway has been described in several species. We searched pubmed for studies reporting the prevention of kidney transplant rejection in nonhuman primates utilizing either anti CD40 or anti CD40L (CD154) treatment. Inclusion of data required treatment with anti CD40 or anti CD154 as monotherapy treatment arms, full text available, studies conducted in nonhuman primate species, the transplant was renal transplantation, sufficient duration of treatment to assess long term rejection, and the reporting of individual graft survival or survival duration. Eleven publications were included in the study. Rejection free survival was calculated using the Kaplan-Meier (KM) life test methods to estimate the survival functions. The 95% CI for the medians was also calculated. A log-rank test was used to test the equality of the survival curves between control and treatment arms (CD40 and CD154). The hazard ratio for CD154 compared to CD40 and 95% CI was calculated using a Cox proportional-hazards model including treatment as the covariate to assess the magnitude of the treatment effect. Both anti CD40 and anti CD154 treatments prevented acute and long term graft rejection. The median (95% CI) rejection free survival was 131 days (84,169 days) in the anti CD40 treated animals and 352 days (173,710 days) in the anti CD154 treated animals. Median survival in the untreated animals was 6 days. The inhibition of transplant rejection was more durable in the anti CD154 group compared to the anti CD40 group after cessation of treatment. The median (95% CI) rejection free survival after cessation of treatment was 60 days (21,80 days) in the anti CD40 treated animals and 230 days (84,552 days) in the anti CD154 treated animals.
Collapse
|