1
|
Byrne GW, McGregor CGA. 2025: status of cardiac xenotransplantation including preclinical models. FRONTIERS IN TRANSPLANTATION 2025; 4:1568910. [PMID: 40302932 PMCID: PMC12037586 DOI: 10.3389/frtra.2025.1568910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025]
Abstract
Xenotransplantation offers an opportunity to radically change the availability of organs for life-saving human transplantation. Great progress has been made in porcine donor genetic engineering to reduce the immunogenicity of pig organs and potentially enhance their resistance to antibody-mediated rejection. There is also growing insight into more effective immune suppression regimens. These advances have improved the duration of cardiac xenograft survival in non-human primates over the last decade and supported the recent approval of the first-in-human clinical use of pig hearts and kidneys for transplantation. This review critically examines preclinical and clinical results in cardiac xenotransplantation. We identify challenges that remain to achieve consistent and durable clinical graft survival. We discuss the relative value of preclinical non-human primate and human decedent transplant models to optimize patient cross-matching, immune suppression, postoperative monitoring, and graft survival.
Collapse
Affiliation(s)
- Guerard W. Byrne
- Department of Surgery, Experimental Surgical Services, University of Minnesota, Minneapolis, MN, United States
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
| | - Christopher G. A. McGregor
- Department of Surgery, Experimental Surgical Services, University of Minnesota, Minneapolis, MN, United States
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
| |
Collapse
|
2
|
Xu H, He X. Developments in kidney xenotransplantation. Front Immunol 2024; 14:1242478. [PMID: 38274798 PMCID: PMC10808336 DOI: 10.3389/fimmu.2023.1242478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 12/13/2023] [Indexed: 01/27/2024] Open
Abstract
The search for kidney xenografts that are appropriate for patients with end-stage renal disease has been ongoing since the beginning of the last century. The major cause of xenograft loss is hyperacute and acute rejection, and this has almost been overcome via scientific progress. The success of two pre-clinical trials of α1,3-galactosyltransferase gene-knockout porcine kidneys in brain-dead patients in 2021 triggered research enthusiasm for kidney xenotransplantation. This minireview summarizes key issues from an immunological perspective: the discovery of key xenoantigens, investigations into key co-stimulatory signal inhibition, gene-editing technology, and immune tolerance induction. Further developments in immunology, particularly immunometabolism, might help promote the long-term outcomes of kidney xenografts.
Collapse
Affiliation(s)
| | - Xiaozhou He
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
3
|
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
|
4
|
Alhejailan RS, Garoffolo G, Raveendran VV, Pesce M. Cells and Materials for Cardiac Repair and Regeneration. J Clin Med 2023; 12:jcm12103398. [PMID: 37240504 DOI: 10.3390/jcm12103398] [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: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
After more than 20 years following the introduction of regenerative medicine to address the problem of cardiac diseases, still questions arise as to the best cell types and materials to use to obtain effective clinical translation. Now that it is definitively clear that the heart does not have a consistent reservoir of stem cells that could give rise to new myocytes, and that there are cells that could contribute, at most, with their pro-angiogenic or immunomodulatory potential, there is fierce debate on what will emerge as the winning strategy. In this regard, new developments in somatic cells' reprogramming, material science and cell biophysics may be of help, not only for protecting the heart from the deleterious consequences of aging, ischemia and metabolic disorders, but also to boost an endogenous regeneration potential that seems to be lost in the adulthood of the human heart.
Collapse
Affiliation(s)
- Reem Saud Alhejailan
- Cell Biology Department, King's Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Gloria Garoffolo
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| | - Vineesh Vimala Raveendran
- Cell Biology Department, King's Faisal Specialist Hospital & Research Center, Riyadh 11564, Saudi Arabia
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| |
Collapse
|
5
|
Sykes M, Sachs DH. Progress in xenotransplantation: overcoming immune barriers. Nat Rev Nephrol 2022; 18:745-761. [PMID: 36198911 DOI: 10.1038/s41581-022-00624-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
A major limitation of organ allotransplantation is the insufficient supply of donor organs. Consequently, thousands of patients die every year while waiting for a transplant. Progress in xenotransplantation that has permitted pig organ graft survivals of years in non-human primates has led to renewed excitement about the potential of this approach to alleviate the organ shortage. In 2022, the first pig-to-human heart transplant was performed on a compassionate use basis, and xenotransplantation experiments using pig kidneys in deceased human recipients provided encouraging data. Many advances in xenotransplantation have resulted from improvements in the ability to genetically modify pigs using CRISPR-Cas9 and other methodologies. Gene editing has the capacity to generate pig organs that more closely resemble those of humans and are hence more physiologically compatible and less prone to rejection. Despite such modifications, immune responses to xenografts remain powerful and multi-faceted, involving innate immune components that do not attack allografts. Thus, the induction of innate and adaptive immune tolerance to prevent rejection while preserving the capacity of the immune system to protect the recipient and the graft from infection is desirable to enable clinical xenotransplantation.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA. .,Department of Surgery, Columbia University, New York, NY, USA. .,Department of Microbiology and Immunology, Columbia University, New York, NY, USA.
| | - David H Sachs
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA. .,Department of Surgery, Columbia University, New York, NY, USA.
| |
Collapse
|
6
|
Chaban R, Cooper DKC, Pierson RN. Pig heart and lung xenotransplantation: Present status. J Heart Lung Transplant 2022; 41:1014-1022. [PMID: 35659792 PMCID: PMC10124776 DOI: 10.1016/j.healun.2022.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/14/2022] [Accepted: 04/24/2022] [Indexed: 11/19/2022] Open
Abstract
The recent pig heart transplant in a patient at the University of Maryland Medical Center has stimulated renewed interest in the xenotransplantation of organs from genetically engineered pigs. The barriers to the use of pigs as sources of organs have largely been overcome by 2 approaches - (1) the deletion of expression of the three known pig carbohydrate xenoantigens against which humans have preformed antibodies, and (2) the transgenic introduction of human 'protective' proteins, such as complement-regulatory proteins. These gene modifications, coupled with immunosuppressive therapy based on blockade of the CD40/CD154 costimulation pathway, have resulted in survival of baboons with life-supporting pig heart grafts for almost 9 months. The initial clinical success at the University of Maryland reinforces encouraging preclinical results. It suggests that pig hearts are likely to provide an effective bridge to an allotransplant, but their utility for destination therapy remains uncertain. Because of additional complex immunobiological problems, the same approach has been less successful in preclinical lung xenograft transplantation, where survival is still measured in days or weeks. The first formal clinical trials of pig heart transplantation may include patients who do not have access to an allotransplant, those with contraindications for mechanical circulatory support, those in need of retransplantation or with a high level of panel-reactive antibodies. Infants with complex congenital heart disease, should also be considered.
Collapse
Affiliation(s)
- Ryan Chaban
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Department of Cardiovascular Surgery, University Hospital of Johannes Gutenberg University, Mainz, Germany.
| | - David K C Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard N Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Pierson RN, Hawthorne WJ. Agnès Marie Azimzadeh, Ph.D – “In Memoriam”. Xenotransplantation 2021. [DOI: 10.1111/xen.12689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Richard N. Pierson
- Department of Surgery Massachusetts General Hospital and Harvard Medical School Boston MA USA
- Center for Transplantation Sciences Massachusetts General Hospital Boston MA USA
| | - Wayne J. Hawthorne
- Centre for Transplant and Renal Research The Westmead Institute for Medical Research Westmead Hospital Sydney NSW Australia
- Department of Surgery Western Clinical School University of Sydney Sydney NSW Australia
| |
Collapse
|
8
|
Galow AM, Goldammer T, Hoeflich A. Xenogeneic and Stem Cell-Based Therapy for Cardiovascular Diseases: Genetic Engineering of Porcine Cells and Their Applications in Heart Regeneration. Int J Mol Sci 2020; 21:ijms21249686. [PMID: 33353186 PMCID: PMC7766969 DOI: 10.3390/ijms21249686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular diseases represent a major health concern worldwide with few therapy options for ischemic injuries due to the limited regeneration potential of affected cardiomyocytes. Innovative cell replacement approaches could facilitate efficient regenerative therapy. However, despite extensive attempts to expand primary human cells in vitro, present technological limitations and the lack of human donors have so far prevented their broad clinical use. Cell xenotransplantation might provide an ethically acceptable unlimited source for cell replacement therapies and bridge the gap between waiting recipients and available donors. Pigs are considered the most suitable candidates as a source for xenogeneic cells and tissues due to their anatomical and physiological similarities with humans. The potential of porcine cells in the field of stem cell-based therapy and regenerative medicine is under intensive investigation. This review outlines the current progress and highlights the most promising approaches in xenogeneic cell therapy with a focus on the cardiovascular system.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Correspondence: ; Tel.: +49-38208-68-723
| | - Tom Goldammer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
- Molecular Biology and Fish Genetics Unit, Faculty of Agriculture and Environmental Sciences, University of Rostock, 18059 Rostock, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology, 18196 Dummerstorf, Germany; (T.G.); (A.H.)
| |
Collapse
|
9
|
Sykes M, Sachs DH. Transplanting organs from pigs to humans. Sci Immunol 2020; 4:4/41/eaau6298. [PMID: 31676497 DOI: 10.1126/sciimmunol.aau6298] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022]
Abstract
The success of organ transplantation is limited by the complications of immunosuppression, by chronic rejection, and by the insufficient organ supply, and thousands of patients die every year while waiting for a transplant. With recent progress in xenotransplantation permitting porcine organ graft survival of months or even years in nonhuman primates, there is renewed interest in its potential to alleviate the organ shortage. Many of these advances are the result of our heightened capacity to modify pigs genetically, particularly with the development of CRISPR-Cas9-based gene editing methodologies. Although this approach allows the engineering of pig organs that are less prone to rejection, the clinical application of xenotransplantation will require the ability to avoid the ravages of a multifaceted attack on the immune system while preserving the capacity to protect both the recipient and the graft from infectious microorganisms. In this review, we will discuss the potential and limitations of these modifications and how the engineering of the graft can be leveraged to alter the host immune response so that all types of immune attack are avoided.
Collapse
Affiliation(s)
- Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, NY, USA. .,Department of Microbiology and Immunology, Columbia University Medical Center, NY, USA.,Department of Surgery, Columbia University Medical Center, NY, USA
| | - David H Sachs
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, NY, USA.,Department of Surgery, Columbia University Medical Center, NY, USA
| |
Collapse
|
10
|
Samy KP, Butler JR, Li P, Cooper DKC, Ekser B. The Role of Costimulation Blockade in Solid Organ and Islet Xenotransplantation. J Immunol Res 2017; 2017:8415205. [PMID: 29159187 PMCID: PMC5660816 DOI: 10.1155/2017/8415205] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/17/2017] [Indexed: 12/17/2022] Open
Abstract
Pig-to-human xenotransplantation offers a potential bridge to the growing disparity between patients with end-stage organ failure and graft availability. Early studies attempting to overcome cross-species barriers demonstrated robust humoral immune responses to discordant xenoantigens. Recent advances have led to highly efficient and targeted genomic editing, drastically altering the playing field towards rapid production of less immunogenic porcine tissues and even the discussion of human xenotransplantation trials. However, as these humoral immune barriers to cross-species transplantation are overcome with advanced transgenics, cellular immunity to these novel xenografts remains an outstanding issue. Therefore, understanding and optimizing immunomodulation will be paramount for successful clinical xenotransplantation. Costimulation blockade agents have been introduced in xenotransplantation research in 2000 with anti-CD154mAb. Most recently, prolonged survival has been achieved in solid organ (kidney xenograft survival > 400 days with anti-CD154mAb, heart xenograft survival > 900 days, and liver xenograft survival 29 days with anti-CD40mAb) and islet xenotransplantation (>600 days with anti-CD154mAb) with the use of these potent experimental agents. As the development of novel genetic modifications and costimulation blocking agents converges, we review their impact thus far on preclinical xenotransplantation and the potential for future application.
Collapse
Affiliation(s)
- Kannan P. Samy
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James R. Butler
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ping Li
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David K. C. Cooper
- Xenotransplantation Program, Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
11
|
Mohiuddin MM, Singh AK, Corcoran PC, Thomas III ML, Clark T, Lewis BG, Hoyt RF, Eckhaus M, Pierson III RN, Belli AJ, Wolf E, Klymiuk N, Phelps C, Reimann KA, Ayares D, Horvath KA. Chimeric 2C10R4 anti-CD40 antibody therapy is critical for long-term survival of GTKO.hCD46.hTBM pig-to-primate cardiac xenograft. Nat Commun 2016; 7:11138. [PMID: 27045379 PMCID: PMC4822024 DOI: 10.1038/ncomms11138] [Citation(s) in RCA: 352] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/23/2016] [Indexed: 12/11/2022] Open
Abstract
Preventing xenograft rejection is one of the greatest challenges of transplantation medicine. Here, we describe a reproducible, long-term survival of cardiac xenografts from alpha 1-3 galactosyltransferase gene knockout pigs, which express human complement regulatory protein CD46 and human thrombomodulin (GTKO.hCD46.hTBM), that were transplanted into baboons. Our immunomodulatory drug regimen includes induction with anti-thymocyte globulin and αCD20 antibody, followed by maintenance with mycophenolate mofetil and an intensively dosed αCD40 (2C10R4) antibody. Median (298 days) and longest (945 days) graft survival in five consecutive recipients using this regimen is significantly prolonged over our recently established survival benchmarks (180 and 500 days, respectively). Remarkably, the reduction of αCD40 antibody dose on day 100 or after 1 year resulted in recrudescence of anti-pig antibody and graft failure. In conclusion, genetic modifications (GTKO.hCD46.hTBM) combined with the treatment regimen tested here consistently prevent humoral rejection and systemic coagulation pathway dysregulation, sustaining long-term cardiac xenograft survival beyond 900 days.
Collapse
Affiliation(s)
| | - Avneesh K. Singh
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, Maryland 20892, USA
| | - Philip C. Corcoran
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, Maryland 20892, USA
| | | | | | - Billeta G. Lewis
- Division of Veterinary Resources, ORS, NIH, Bethesda, Maryland 20892, USA
| | - Robert F. Hoyt
- Leidos Biomedical Research, Inc., Bethesda, Maryland 20892, USA
| | - Michael Eckhaus
- Division of Veterinary Resources, ORS, NIH, Bethesda, Maryland 20892, USA
| | | | - Aaron J. Belli
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts 02126, USA
| | - Eckhard Wolf
- Ludwig Maximilian University, Munich 81377, Germany
| | | | | | - Keith A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts 02126, USA
| | | | - Keith A. Horvath
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, Maryland 20892, USA
| |
Collapse
|
12
|
Lee W, Satyananda V, Iwase H, Tanaka T, Miyagawa Y, Long C, Ayares D, Cooper DKC, Hara H. In vitro testing of an anti-CD40 monoclonal antibody, clone 2C10, in primates and pigs. Transpl Immunol 2015; 33:185-91. [PMID: 26458513 PMCID: PMC4648655 DOI: 10.1016/j.trim.2015.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND The CD40/CD154 and CD28/B7 pathways are important in allo- and xeno-transplantation. Owing to the thrombotic complications of anti-CD154mAb, anti-CD40mAb has emerged as a promising inhibitor of costimulation. Various clones of anti-CD40mAb have been developed against primate species, e.g., clone 2C10 against rhesus monkeys. We have compared the in vitro efficacy of 2C10 to prevent a T cell response in primates and pigs. METHODS The binding of 2C10 to antigen-presenting cells (PBMCs [B cells]) of humans, rhesus and cynomolgus monkeys, baboons, and pigs was measured by flow cytometry, and was also tested indirectly by a blocking assay. The functional capacity of 2C10 was tested by mixed lymphocyte reaction (MLR) with polyclonal stimulation by phytohemagglutinin (PHA) and also with wild-type pig aortic endothelial cells (pAECs) as stimulators. RESULTS There was a significant reduction in binding of 2C10 to baboon PBMCs compared to rhesus, cynomolgus, and human PBMCs, and minimal binding to pig PBMCs. The blocking assay confirmed that the binding of 2C10 was significantly lower to baboon PBMCs when compared to the other primate species tested. The functional assay with PHA showed significantly reduced inhibition of PBMC proliferation in humans, cynomolgus monkeys, and baboons compared to rhesus monkeys, which was confirmed on MLR with pAECs. CONCLUSIONS Since both the binding and functional activity of 2C10 in the baboon is lower than in rhesus monkeys, in vivo treatment using 2C10 in the baboon might require a higher dose or more frequent administration in comparison to rhesus monkeys. It may also be beneficial to develop species-specific clones of anti-CD40mAb.
Collapse
Affiliation(s)
- Whayoung Lee
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vikas Satyananda
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hayato Iwase
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takayuki Tanaka
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuko Miyagawa
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cassandra Long
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hidetaka Hara
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Byrne GW, McGregor CGA, Breimer ME. Recent investigations into pig antigen and anti-pig antibody expression. Int J Surg 2015; 23:223-228. [PMID: 26306769 DOI: 10.1016/j.ijsu.2015.07.724] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 07/22/2015] [Accepted: 07/26/2015] [Indexed: 01/17/2023]
Abstract
Genetic engineering of donor pigs to eliminate expression of the dominant xenogeneic antigen galactose α1,3 galactose (Gal) has created a sea change in the immunobiology of xenograft rejection. Antibody mediated xenograft rejection of GGTA-1 α-galactosyltransferase (GTKO) deficient organs is now directed to a combination of non-Gal pig protein and carbohydrate antigens. Glycan analysis of GTKO tissues identified no new neo-antigens but detected high levels of N-acetylneuraminic acid (Neu5Gc) modified glycoproteins and glycolipids. Humans produce anti-Neu5Gc antibody and in very limited clinical studies sometimes show an induced anti-Neu5Gc antibody response after challenge with pig tissue. The pathogenicity of anti-Neu5Gc antibody in xenotransplantation is not clear however as non-human transplant models, critical for modelling anti-Gal immunity, do not produce anti-Neu5Gc antibody. Antibody induced after xenotransplantation in non-human primates is directed to an array of pig endothelial cells proteins and to a glycan produced by the pig B4GALNT2 gene. We anticipate that immune suppression will significantly affect the T-cell dependent and independent specificity of an induced antibody response and that donor pigs deficient in synthesis of multiple xenogeneic glycans will be important to future studies.
Collapse
Affiliation(s)
- Guerard W Byrne
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA; Institute of Cardiovascular Science, University College London, London WC1E 6JF, UK.
| | - Christopher G A McGregor
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA; Institute of Cardiovascular Science, University College London, London WC1E 6JF, UK
| | - Michael E Breimer
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
14
|
Iwase H, Ekser B, Satyananda V, Zhou H, Hara H, Bajona P, Wijkstrom M, Bhama JK, Long C, Veroux M, Wang Y, Dai Y, Phelps C, Ayares D, Ezzelarab MB, Cooper DKC. Initial in vivo experience of pig artery patch transplantation in baboons using mutant MHC (CIITA-DN) pigs. Transpl Immunol 2015; 32:99-108. [PMID: 25687023 PMCID: PMC4368496 DOI: 10.1016/j.trim.2015.02.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND In the pig-to-nonimmunosuppressed baboon artery patch model, a graft from an α1,3-galactosyltransferase gene-knockout pig transgenic for human CD46 (GTKO/CD46) induces a significant adaptive immune response (elicited anti-pig antibody response, increase in T cell proliferation on MLR, cellular infiltration of the graft), which is effectively prevented by anti-CD154mAb-based therapy. METHODS As anti-CD154mAb is currently not clinically applicable, we evaluated whether it could be replaced by CD28/B7 pathway blockade or by blockade of both pathways (using belatacept + anti-CD40mAb [2C10R4]). We further investigated whether a patch from a GTKO/CD46 pig with a mutant human MHC class II transactivator (CIITA-DN) gene would allow reduction in the immunosuppressive therapy administered. RESULTS When grafts from GTKO/CD46 pigs were transplanted with blockade of both pathways, a minimal or insignificant adaptive response was documented. When a GTKO/CD46/CIITA-DN graft was transplanted, but no immunosuppressive therapy was administered, a marked adaptive response was documented. In the presence of CD28/B7 pathway blockade (abatacept or belatacept), there was a weak adaptive response that was diminished when compared with that to a GTKO/CD46 graft. Blockade of both pathways prevented an adaptive response. CONCLUSION Although expression of the mutant MHC CIITA-DN gene was associated with a reduced adaptive immune response when immunosuppressive therapy was inadequate, when blockade of both the CD40/CD154 and CD28/B7 pathways was present, the response even to a GTKO/CD46 graft was suppressed. This was confirmed after GTKO/CD46 heart transplantation in baboons.
Collapse
Affiliation(s)
- H Iwase
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - B Ekser
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, Transplantation and Advanced Technologies, Vascular Surgery and Organ Transplant Unit, University Hospital of Catania, Catania, Italy
| | - V Satyananda
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - H Zhou
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA; Center for Kidney Transplantation, Second Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - H Hara
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - P Bajona
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Wijkstrom
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - J K Bhama
- Department of Cardiac Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - C Long
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Veroux
- Department of Surgery, Transplantation and Advanced Technologies, Vascular Surgery and Organ Transplant Unit, University Hospital of Catania, Catania, Italy
| | - Y Wang
- Center for Kidney Transplantation, Second Affiliated Hospital of the University of South China, Hengyang, Hunan, China
| | - Y Dai
- Revivicor, Blacksburg, VA, USA
| | | | | | - M B Ezzelarab
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - D K C Cooper
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
De Salvatore S, Segreto A, Chiusaroli A, Congiu S, Bizzarri F. Role of xenotransplantation in cardiac transplantation. J Card Surg 2014; 30:111-6. [PMID: 25345720 DOI: 10.1111/jocs.12454] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
This review will discuss the history and development of the field of genetic modification, up to the most recent scientific discoveries, and will also consider the current uses of genetic therapy.
Collapse
Affiliation(s)
- Sergio De Salvatore
- Department of Science and Medical-Surgical Biotechnologies, Cardiac Surgery Unit, Universita' degli Studi di Roma "Sapienza", Latina, Italy
| | | | | | | | | |
Collapse
|
16
|
Azimzadeh AM, Byrne GW, Ezzelarab M, Welty E, Braileanu G, Cheng X, Robson SC, McGregor CGA, Cooper DKC, Pierson RN. Development of a consensus protocol to quantify primate anti-non-Gal xenoreactive antibodies using pig aortic endothelial cells. Xenotransplantation 2014; 21:555-66. [PMID: 25176173 DOI: 10.1111/xen.12125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 05/30/2014] [Indexed: 01/05/2023]
Abstract
BACKGROUND Scientists working in the field of xenotransplantation do not employ a uniform method to measure and report natural and induced antibody responses to non-Galα(1,3)Gal (non-Gal) epitopes. Such humoral responses are thought to be particularly pathogenic after transplantation of vascularized GalTKO pig organs and having a more uniform assay and reporting format would greatly facilitate comparisons between laboratories. METHODS Flow cytometry allows examination of antibody reactivity to intact antigens in their natural location and conformation on cell membranes. We have established a simple and reproducible flow cytometric assay to detect antibodies specific for non-Gal pig antigens using primary porcine aortic endothelial cells (pAECs) and cell culture-adapted pAEC cell lines generated from wild type and α1,3galactosyl transferase knockout (GalTKO) swine. RESULTS The consensus protocol we propose here is based on procedures routinely used in four xenotransplantation centers and was independently evaluated at three sites using shared cells and serum samples. Our observation support use of the cell culture-adapted GalTKO pAEC KO:15502 cells as a routine method to determine the reactivity of anti-non-Gal antibodies in human and baboon serum. CONCLUSIONS We have developed an assay that allows the detection of natural and induced non-Gal xenoreactive antibodies present in human or baboon serum in a reliable and consistent manner. This consensus assay and format for reporting the data should be accessible to laboratories and will be useful for assessing experimental results between multiple research centers. Adopting this assay and format for reporting the data should facilitate the detection, monitoring, and detailed characterization of non-Gal antibody responses.
Collapse
Affiliation(s)
- Agnes M Azimzadeh
- Division of Cardiac Surgery, University of Maryland and VAMC Baltimore, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Cooper DKC, Satyananda V, Ekser B, van der Windt DJ, Hara H, Ezzelarab MB, Schuurman HJ. Progress in pig-to-non-human primate transplantation models (1998-2013): a comprehensive review of the literature. Xenotransplantation 2014; 21:397-419. [PMID: 25176336 DOI: 10.1111/xen.12127] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/03/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND The pig-to-non-human primate model is the standard choice for in vivo studies of organ and cell xenotransplantation. In 1998, Lambrigts and his colleagues surveyed the entire world literature and reported all experimental studies in this model. With the increasing number of genetically engineered pigs that have become available during the past few years, this model is being utilized ever more frequently. METHODS We have now reviewed the literature again and have compiled the data we have been able to find for the period January 1, 1998 to December 31, 2013, a period of 16 yr. RESULTS The data are presented for transplants of the heart (heterotopic and orthotopic), kidney, liver, lung, islets, neuronal cells, hepatocytes, corneas, artery patches, and skin. Heart, kidney, and, particularly, islet xenograft survival have increased significantly since 1998. DISCUSSION The reasons for this are briefly discussed. A comment on the limitations of the model has been made, particularly with regard to those that will affect progression of xenotransplantation toward the clinic.
Collapse
Affiliation(s)
- David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Mohiuddin MM, Singh AK, Corcoran PC, Hoyt RF, Thomas ML, Ayares D, Horvath KA. Genetically engineered pigs and target-specific immunomodulation provide significant graft survival and hope for clinical cardiac xenotransplantation. J Thorac Cardiovasc Surg 2014; 148:1106-13; discussion 1113-4. [PMID: 24998698 PMCID: PMC4135017 DOI: 10.1016/j.jtcvs.2014.06.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Cardiac transplantation and available mechanical alternatives are the only possible solutions for end-stage cardiac disease. Unfortunately, because of the limited supply of human organs, xenotransplantation may be the ideal method to overcome this shortage. We have recently seen significant prolongation of heterotopic cardiac xenograft survival from 3 to 12 months and beyond. METHODS Hearts from genetically engineered piglets that were alpha 1-3 galactosidase transferase knockout and expressed the human complement regulatory gene, CD46 (groups A-C), and the human thrombomodulin gene (group D) were heterotropically transplanted in baboons treated with antithymocyte globulin, cobra venom factor, anti-CD20 antibody, and costimulation blockade (anti-CD154 antibody [clone 5C8]) in group A, anti-CD40 antibody (clone 3A8; 20 mg/kg) in group B, clone 2C10R4 (25 mg/kg) in group C, or clone 2C10R4 (50 mg/kg) in group D, along with conventional nonspecific immunosuppressive agents. RESULTS Group A grafts (n = 8) survived for an average of 70 days, with the longest survival of 236 days. Some animals in this group (n = 3) developed microvascular thrombosis due to platelet activation and consumption, which resulted in spontaneous hemorrhage. The median survival time was 21 days in group B (n = 3), 80 days in group C (n = 6), and more than 200 days in group D (n = 5). Three grafts in group D are still contracting well, with the longest ongoing graft survival surpassing the 1-year mark. CONCLUSIONS Genetically engineered pig hearts (GTKOhTg.hCD46.hTBM) with modified targeted immunosuppression (anti-CD40 monoclonal antibody) achieved long-term cardiac xenograft survival. This potentially paves the way for clinical xenotransplantation if similar survival can be reproduced in an orthotopic transplantation model.
Collapse
Affiliation(s)
- Muhammad M Mohiuddin
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md.
| | - Avneesh K Singh
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Philip C Corcoran
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Robert F Hoyt
- Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc, Frederick National Laboratory, Frederick, Md
| | - Marvin L Thomas
- Division of Veterinary Resources, National Institutes of Health, Bethesda, Md
| | | | - Keith A Horvath
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| |
Collapse
|
19
|
Plege-Fleck A, Lieke T, Römermann D, Düvel H, Hundrieser J, Buermann A, Kraus L, Klempnauer J, Schwinzer R. Pig to rat cell transplantation: reduced cellular and antibody responses to xenografts overexpressing PD-L1. Xenotransplantation 2014; 21:533-42. [DOI: 10.1111/xen.12121] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/28/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Annegret Plege-Fleck
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Thorsten Lieke
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Dorothee Römermann
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Heike Düvel
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Joachim Hundrieser
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Anna Buermann
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Lilli Kraus
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Jürgen Klempnauer
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Reinhard Schwinzer
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| |
Collapse
|
20
|
Mohiuddin MM, Singh AK, Corcoran PC, Hoyt RF, Thomas ML, Lewis BGT, Eckhaus M, Dabkowski NL, Belli AJ, Reimann KA, Ayares D, Horvath KA. Role of anti-CD40 antibody-mediated costimulation blockade on non-Gal antibody production and heterotopic cardiac xenograft survival in a GTKO.hCD46Tg pig-to-baboon model. Xenotransplantation 2014; 21:35-45. [PMID: 24164510 PMCID: PMC5603077 DOI: 10.1111/xen.12066] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 09/16/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recently, we have shown that an immunosuppression regimen including costimulation blockade via anti-CD154 antibody significantly prolongs the cardiac xenograft survival in a GTKO.hCD46Tg pig-to-baboon heterotopic xenotransplantation model. Unfortunately, many coagulation disorders were observed with the use of anti-CD154 antibody, and recipient survival was markedly reduced by these complications. MATERIAL AND METHODS In this experiment, we replaced anti-CD154 antibody with a more clinically acceptable anti-CD40 antibody while keeping the rest of the immunosuppressive regimen and the donor pig genetics the same. This was carried out to evaluate the antibody's role in xenograft survival and prevention of coagulopathies. Two available clones of anti-CD40 antibody were tested. One mouse anti-human CD40 antibody, (clone 3A8), activated B lymphocytes in vitro and only modestly suppressed antibody production in vivo. Whereas a recombinant mouse non-human primate chimeric raised against macaque CD40, (clone 2C10R4), blocked B-cell activation in vitro and completely blocked antibody production in vivo. RESULTS The thrombotic complications seen with anti-CD154 antibody were effectively avoided but the graft survival, although extended, was not as prolonged as observed with anti-CD154 antibody treatment. The longest survival for the 3A8 antibody group was 27 days, and the longest graft survival in the 2C10R4 antibody group was 146 days. All of the grafts except two rejected and were explanted. Only two recipient baboons had to be euthanized due to unrelated complications, and the rest of the baboons remained healthy throughout the graft survival period or after graft explantation. In contrast to our anti-CD 154 antibody-treated baboons, the non-Gal antibody levels started to rise after B cells made their appearance around 8 weeks post-transplantation. CONCLUSIONS Anti-CD40 antibody at the current dose does not induce any coagulopathies but while effective, had reduced efficacy to induce similar long-term graft survival as with anti-CD154 antibody perhaps due to ineffective control of B-cell function and antibody production at the present dose. More experiments are required to determine antibody affinity and effective dose for inducing long-term cardiac xenograft survival.
Collapse
Affiliation(s)
| | - Avneesh K. Singh
- Cardiothoracic Surgery Research Laboratory, NHLBI, NIH, Bethesda, MD
| | | | | | | | | | | | | | - Aaron J. Belli
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - Keith A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | | | - Keith A. Horvath
- Cardiothoracic Surgery Research Laboratory, NHLBI, NIH, Bethesda, MD
| |
Collapse
|
21
|
Byrne GW, Azimzadeh AM, Ezzelarab M, Tazelaar HD, Ekser B, Pierson RN, Robson SC, Cooper DKC, McGregor CGA. Histopathologic insights into the mechanism of anti-non-Gal antibody-mediated pig cardiac xenograft rejection. Xenotransplantation 2013; 20:292-307. [PMID: 25098626 PMCID: PMC4126170 DOI: 10.1111/xen.12050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/31/2013] [Indexed: 01/13/2023]
Abstract
The histopathology of cardiac xenograft rejection has evolved over the last 20 yr with the development of new modalities for limiting antibody-mediated injury, advancing regimens for immune suppression, and an ever-widening variety of new donor genetics. These new technologies have helped us progress from what was once an overwhelming anti-Gal-mediated hyperacute rejection to a more protracted anti-Gal-mediated vascular rejection to what is now a more complex manifestation of non-Gal humoral rejection and coagulation dysregulation. This review summarizes the changing histopathology of Gal- and non-Gal-mediated cardiac xenograft rejection and discusses the contributions of immune-mediated injury, species-specific immune-independent factors, transplant and therapeutic procedures, and donor genetics to the overall mechanism(s) of cardiac xenograft rejection.
Collapse
Affiliation(s)
- Guerard W Byrne
- Institute of Cardiovascular Science, University College London, London, UK; Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mohiuddin MM, Corcoran PC, Singh AK, Azimzadeh A, Hoyt RF, Thomas ML, Eckhaus MA, Seavey C, Ayares D, Pierson RN, Horvath KA. B-cell depletion extends the survival of GTKO.hCD46Tg pig heart xenografts in baboons for up to 8 months. Am J Transplant 2012; 12:763-71. [PMID: 22070772 PMCID: PMC4182960 DOI: 10.1111/j.1600-6143.2011.03846.x] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Xenotransplantation of genetically modified pig organs offers great potential to address the shortage of human organs for allotransplantation. Rejection in Gal knockout (GTKO) pigs due to elicited non-Gal antibody response required further genetic modifications of donor pigs and better control of the B-cell response to xenoantigens. We report significant prolongation of heterotopic alpha Galactosyl transferase "knock-out" and human CD46 transgenic (GTKO.hCD46Tg) pig cardiac xenografts survival in specific pathogen free baboons. Peritransplant B-cell depletion using 4 weekly doses of anti-CD20 antibody in the context of an established ATG, anti-CD154 and MMF-based immunosuppressive regimen prolonged GTKO.hCD46Tg graft survival for up to 236 days (n = 9, median survival 71 days and mean survival 94 days). B-cell depletion persisted for over 2 months, and elicited anti-non-Gal antibody production remained suppressed for the duration of graft follow-up. This result identifies a critical role for B cells in the mechanisms of elicited anti-non-Gal antibody and delayed xenograft rejection. Model-related morbidity due to variety of causes was seen in these experiments, suggesting that further therapeutic interventions, including candidate genetic modifications of donor pigs, may be necessary to reduce late morbidity in this model to a clinically manageable level.
Collapse
Affiliation(s)
- MM Mohiuddin
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, MD
| | - PC Corcoran
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, MD
| | - AK Singh
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, MD
| | - A Azimzadeh
- University of Maryland Medical Center, Baltimore, MD
| | - RF Hoyt
- LAMS, NHLBI, NIH, Bethesda, MD
| | | | | | - C Seavey
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, MD
| | | | - RN Pierson
- University of Maryland Medical Center, Baltimore, MD
| | - KA Horvath
- Cardiothoracic Surgery Research Program, NHLBI, NIH, Bethesda, MD
| |
Collapse
|
23
|
Pierson RN, Dorling A, Ayares D, Rees MA, Seebach JD, Fishman JA, Hering BJ, Cooper DKC. Current status of xenotransplantation and prospects for clinical application. Xenotransplantation 2009; 16:263-80. [PMID: 19796067 PMCID: PMC2866107 DOI: 10.1111/j.1399-3089.2009.00534.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Xenotransplantation is one promising approach to bridge the gap between available human cells, tissues, and organs and the needs of patients with diabetes or end-stage organ failure. Based on recent progress using genetically modified source pigs, improving results with conventional and experimental immunosuppression, and expanded understanding of residual physiologic hurdles, xenotransplantation appears likely to be evaluated in clinical trials in the near future for some select applications. This review offers a comprehensive overview of known mechanisms of xenograft injury, a contemporary assessment of preclinical progress and residual barriers, and our opinions regarding where breakthroughs are likely to occur.
Collapse
Affiliation(s)
- Richard N Pierson
- Division of Cardiac Surgery, Department of Surgery, University of Maryland, Baltimore VAMC, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Pierson RN. Antibody-mediated xenograft injury: mechanisms and protective strategies. Transpl Immunol 2009; 21:65-9. [PMID: 19376229 PMCID: PMC2695451 DOI: 10.1016/j.trim.2009.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Revised: 12/09/2008] [Accepted: 03/25/2009] [Indexed: 11/28/2022]
Abstract
The use of porcine organs for clinical transplantation is a promising potential solution to the shortage of human organs. Preformed anti-pig antibody is the primary cause of hyperacute rejection, while elicited antibody can contribute to subsequent "delayed" xenograft rejection. This article will review recent progress to overcome antibody mediated xenograft rejection, through modification of the host immunity and use of genetically engineered pig organs.
Collapse
Affiliation(s)
- Richard N Pierson
- Department of Surgery, University of Maryland School of Medicine and Baltimore VAMC, Baltimore, MD 21201, USA.
| |
Collapse
|
25
|
Ezzelarab M, Garcia B, Azimzadeh A, Sun H, Lin CC, Hara H, Kelishadi S, Zhang T, Lin YJ, Tai HC, Wagner R, Thacker J, Murase N, McCurry K, Barth RN, Ayares D, Pierson RN, Cooper DKC. The innate immune response and activation of coagulation in alpha1,3-galactosyltransferase gene-knockout xenograft recipients. Transplantation 2009; 87:805-12. [PMID: 19300181 PMCID: PMC4135362 DOI: 10.1097/tp.0b013e318199c34f] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The role of the innate immune system in the development of thrombotic microangiopathy (TM) after alpha1,3-galactosyltransferase gene-knockout (GTKO) pig organ transplantation in primates is uncertain. METHODS Twelve organs (nine hearts, three kidneys) from GTKO pigs were transplanted into baboons that received no immunosuppressive therapy, partial regimens, or a full regimen based on costimulation blockade. After graft failure, histologic and immunohistologic examinations were carried out. RESULTS Graft survival of less than 1 day was prolonged to 2 to 12 days with partial regimens (acute humoral xenograft rejection) and to 5 and 8 weeks with the full regimen (TM). Clinical or laboratory features of consumptive coagulopathy occurred in 7 of 12 baboons. Immunohistochemistry demonstrated IgM, IgG, and complement deposition in most cases. Histopathology demonstrated neutrophil and macrophage infiltrates, intravascular fibrin deposition, and platelet aggregation (TM). Grafts showed expression of primate tissue factor (TF), with increased mRNA levels, and TF was also expressed on baboon macrophages/monocytes infiltrating the graft. CONCLUSIONS Our data suggest that (1) irrespective of the presence or absence of the adaptive immune response, early or late xenograft rejection is associated with activation of the innate immune system; and (2) porcine endothelial cell activation and primate TF expression by recipient innate immune cells may both contribute to the development of TM.
Collapse
Affiliation(s)
- Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tredget EB, Arefanian H, Gill RG, Rajotte RV, Rayat GR. Monotherapy with anti-LFA-1 monoclonal antibody promotes long-term survival of rat islet xenografts. Cell Transplant 2008; 17:599-608. [PMID: 18819248 DOI: 10.3727/096368908786092757] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Previously we demonstrated that anti-LFA-1 monoclonal (mAb) could promote long-term survival of discordant porcine islet xenografts in mice. The aim of this study, therefore, was to determine whether a shortterm administration of anti-LFA-1 mAb would promote long-term survival of concordant rat islet xenografts in mice, and whether combining short-term administration of anti-LFA-1 mAb therapy with an immunosuppressive drug, rapamycin, would facilitate islet xenograft survival. Streptozotocin-induced diabetic BALB/c mice were transplanted with 500 Wistar-Furth rat islets under the kidney capsule and were either left untreated or treated with short-term administration of rapamycin (0.2 mg/kg) alone, anti-LFA-1 mAb (0.2 mg/ dose) alone, or a combination of rapamycin and anti-LFA-1 mAb using the same doses. All untreated mice rejected their grafts by 24 days posttransplantation with a mean graft survival time of 18.8 +/- 2.5 days posttransplantation (n = 5). All mice treated with rapamycin alone had prolonged islet graft survival but eventually rejected their islet grafts by 81 days posttransplantation. In contrast, the majority of the mice (27/ 28) treated with anti-LFA-1 mAb alone maintained long-term normoglycemia (>100 days). Rapamycin in combination with anti-LFA-1 mAb proved equally effective with 29 of 30 mice maintaining normoglycemia for more than 100 days posttransplantation. Low levels of mouse anti-rat antibodies, as well as a decrease in the degree of mononuclear cell infiltration of the islet graft, closely correlated with long-term islet xenograft survival. These results demonstrate that monotherapy with anti-LFA-1 mAb is highly effective in promoting long-term survival of rat islet xenografts and that combination of anti-LFA-1 mAb with rapamycin does not facilitate nor abrogate the induction of long-term xenograft survival by anti-LFA-1 mAb therapy in BALB/c mice. Our study indicates that immunomodulation through mAb therapy could form a significant component of future antirejection therapies in clinical islet xenotransplantation.
Collapse
Affiliation(s)
- Eric B Tredget
- Surgical-Medical Research Institute, Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
27
|
Guillonneau C, Séveno C, Dugast AS, Li XL, Renaudin K, Haspot F, Usal C, Veziers J, Anegon I, Vanhove B. Anti-CD28 antibodies modify regulatory mechanisms and reinforce tolerance in CD40Ig-treated heart allograft recipients. THE JOURNAL OF IMMUNOLOGY 2008; 179:8164-71. [PMID: 18056359 DOI: 10.4049/jimmunol.179.12.8164] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Blockade of CD40-CD40 ligand (CD40L) costimulation has been shown to synergize with that of CTLA4/CD28-B7 to promote transplant tolerance. To date, however, CD28-B7 interactions have been prevented using B7-blocking reagents like CTLA4-Ig that inhibit CD28-B7 together with CTLA4-B7 interactions. In this study, we have tested anti-CD28 Abs to prevent selectively CD28-B7 interactions while preserving CTLA4-B7 in addition to CD40-CD40L blockade. In the LEW.1W to LEW.1A rat combination, interfering with CD40-CD40L interactions by CD40Ig administration through gene transfer resulted in indefinite heart allograft survival due to the appearance of clonotypic CD8+CD45RClow regulatory T cells that were capable of transferring the tolerant state to naive animals. However, cardiac transplants in these recipients systematically developed chronic rejection lesions. Whereas anti-CD28 Ab monotherapy only delayed acute rejection and failed to induce tolerance, coadministration of anti-CD28 Abs and CD40Ig resulted in the long-term acceptation of allografts without chronic rejection lesions in 60% of the recipients, reduced the level of intragraft mRNA transcripts for cytokines and immune factors, and fully abrogated alloantibody production. In addition, the nature of regulatory cells was modified: the CD8+CD45RClow clonotypic T cells described in the CD40Ig-treated animals could not be found in cotreated animals, and the other CD8+CD45RClow cells had no regulatory activity and a different cytokine expression profile. Instead, in cotreated recipients we found IDO-dependent non-T cells with regulatory activity in vitro. Thus, the addition of a short-term anti-CD28 treatment with CD40Ig resulted in decreased heart allograft chronic rejection lesions, complete inhibition of Ab production, and modified regulatory mechanisms.
Collapse
Affiliation(s)
- Carole Guillonneau
- Institut National de la Santé et de la Recherche Médicale, Unité 643, Centre Hospitalier de l'Université Nantes, Institut de Transplantation et de Recherche en Transplantation, Université de Nantes, Faculté de Médecine, Nantes, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhao GH, Sun HZ, Zhong XP, Zhang JX, Xu GY, Zhao N, Cheng Y, Zhang JL, Liu YF. Preventive effect of pcDNA3-CTLA4Ig plasmid transfection on islet allograft rejection in mice. Shijie Huaren Xiaohua Zazhi 2007; 15:1596-1601. [DOI: 10.11569/wcjd.v15.i14.1596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the effect of pcDNA3-CTLA4Ig plasmid on islet allograft rejection in mice.
METHODS: C57BL/6 mice were randomly divided into control group, blank group and experiment group (transfected with pcDNA3-CTLA4Ig plasmid). The level of serum CTLA4Ig was detected by Western blot on day 5 after transfection. Blood glucose was examined after operation every other day. Immunohistochemical staining was used to determine the expression of insulin. 3H-thymidine incorporation was performed to detect the ability of T lymphocyte increment and the levels of CD4+ and CD8+ T lymphocytes were detected by flow cytometry on day 7 after transplantation.
RESULTS: Five days after transfection, Western blot demonstrated serum expression of CTLA4Ig, and the transfection efficacy was 27.50%. Blood glucose maintained at the normal level for a longer period of time in the experiment group, and on day 7, the proliferation of lymphocytes was markedly decreased (P < 0.05). The levels of CD4+ and CD8+ T lymphocytes were significantly lower in the experiment group than those in the control and blank group (CD4+: 14.38% ± 0.84% vs 20.56% ± 0.68%, 21.04% ± 1.14%, P < 0.05; CD8+: 14.77% ± 0.92% vs 24.63% ± 1.30%, 23.84% ± 1.21%, P < 0.05), and the intensity of immunohistochemical staining was also stronger.
CONCLUSION: CTLA4 gene can be transferred into mouse muscular cells by cation liposome and express its products in the serum, which leads to the inhibition of islet allograft rejection by the blockage of B7/CD28 signal pathway.
Collapse
|
29
|
|
30
|
Xenotransplantation of thoracic organs. Curr Opin Organ Transplant 2007. [DOI: 10.1097/mot.0b013e32809876fc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Abstract
Mohiuddin discusses the lessons learned from large animal xenograft models and why the immunological barrier is still the most important hurdle preventing clinical xenotransplantation of organs.
Collapse
Affiliation(s)
- Muhammad M Mohiuddin
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America.
| |
Collapse
|
32
|
Zahorsky-Reeves JL, Kearns-Jonker MK, Lam TT, Jackson JR, Morris RE, Starnes VA, Cramer DV. The xenoantibody response and immunoglobulin gene expression profile of cynomolgus monkeys transplanted with hDAF-transgenic porcine hearts. Xenotransplantation 2007; 14:135-44. [PMID: 17381688 DOI: 10.1111/j.1399-3089.2007.00381.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Recent work has indicated a role for anti-Gal alpha 1-3Gal (Gal) and anti-non-Gal xenoantibodies in the primate humoral rejection response against human-decay accelerating factor (hDAF) transgenic pig organs. Our laboratory has shown that anti-porcine xenograft antibodies in humans and non-human primates are encoded by a small number of germline IgV(H) progenitors. In this study, we extended our analysis to identify the IgV(H) genes encoding xenoantibodies in immunosuppressed cynomolgus monkeys (Macaca fascicularis) transplanted with hDAF-transgenic pig organs. METHODS Three immunosuppressed monkeys underwent heterotopic heart transplantation with hDAF porcine heart xenografts. Two of three animals were given GAS914, a poly-L-lysine derivative shown to bind to anti-Gal xenoantibodies and neutralize them. One animal rejected its heart at post-operative day (POD) 39; a second animal rejected the transplanted heart at POD 78. The third monkey was euthanized on POD 36 but the heart was not rejected. Peripheral blood leukocytes (PBL) and serum were obtained from each animal before and at multiple time points after transplantation. We analyzed the immune response by enzyme-linked immunosorbent assay (ELISA) to confirm whether anti-Gal or anti-non-Gal xenoantibodies were induced after graft placement. Immunoglobulin heavy-chain gene (V(H)) cDNA libraries were then produced and screened. We generated soluble single-chain antibodies (scFv) to establish the binding specificity of the cloned immunoglobulin genes. RESULTS Despite immunosuppression, which included the use of the polymer GAS914, the two animals that rejected their hearts showed elevated levels of cytotoxic anti-pig red blood cell (RBC) antibodies and anti-pig aortic endothelial cell (PAEC) antibodies. The monkey that did not reject its graft showed a decline in serum anti-RBC, anti-PAEC, and anti-Gal xenoantibodies when compared with pre-transplant levels. A V(H)3 family gene with a high level of sequence similarity to an allele of V(H)3-11, designated V(H)3-11(cyno), was expressed at elevated levels in the monkey that was not given GAS914 and whose graft was not rejected until POD 78. IgM but not IgG xenoantibodies directed at N-acetyl lactosamine (a precursor of the Gal epitope) were also induced in this animal. We produced soluble scFv from this new gene to determine whether this antibody could bind to the Gal carbohydrate, and demonstrated that this protein was capable of blocking the binding of human serum xenoantibody to Gal oligosaccharide, as had previously been shown with human V(H)3-11 scFv. CONCLUSIONS DAF-transgenic organs transplanted into cynomolgus monkeys induce anti-Gal and anti-non-Gal xenoantibody responses mediated by both IgM and IgG xenoantibodies. Anti-non-Gal xenoantibodies are induced at high levels in animals treated with GAS914. Antibodies that bind to the Gal carbohydrate and to N-acetyl lactosamine are induced in the absence of GAS914 treatment. The animal whose heart remained beating for 78 days demonstrated increased usage of an antibody encoded by a germline progenitor that is structurally related, but distinct from IGHV311. This antibody binds to the Gal carbohydrate but does not induce the rapid rejection of the xenograft when expressed at high levels as early as day 8 post-transplantation.
Collapse
Affiliation(s)
- Joanne L Zahorsky-Reeves
- Cardiothoracic Surgery Research, The Saban Research Institute of Childrens Hospital Los Angeles, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Wu G, Pfeiffer S, Schröder C, Zhang T, Nguyen BN, Kelishadi S, Atkinson JB, Schuurman HJ, White DJG, Azimzadeh AM, Pierson RN. Coagulation cascade activation triggers early failure of pig hearts expressing human complement regulatory genes. Xenotransplantation 2007; 14:34-47. [PMID: 17214703 DOI: 10.1111/j.1399-3089.2006.00362.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Hyperacute rejection (HAR) and early graft failure (EGF) have been described in a minority of pig-to-baboon heart transplants using organs transgenic for human complement regulatory proteins (hCRP). Here we investigate the role of coagulation cascade activation in the pathogenesis of HAR and EGF in a consecutive series where a high incidence of these outcomes was observed. METHODS Twenty-eight naïve wild-caught Papio anubis baboons received heterotopic heart transplants from pigs transgenic for hDAF (n = 23) or hMCP (n = 5). Immunosuppression consisted of cyclosporine A, cyclophosphamide and MMF (n = 18) or anti-CD154 mAb (IDEC-131) and ATG (n = 10). Eleven received anti-Gal carbohydrates (GAS914, n = 8, or NEX1285, n = 3), of which four also underwent extracorporeal immunoadsorption (EIA), and 12 also received pharmacologic complement inhibitors (C1 INH, n = 9, or APT070, n = 3). RESULTS Excluding one technical failure, 14 of 27 transplants (11 hDAF, 3 hMCP) exhibited either HAR (n = 10) or EGF (n = 4). Surprisingly, neither complement inhibition (with C1 INH or APT070) nor anti-Gal antibody depletion with GAS914, NEX1285, or additional EIA consistently prevented HAR or EGF despite low or undetectable complement deposition. Strikingly, most grafts with HAR/EGF exhibited prominent fibrinogen and platelet deposition associated with systemic coagulation cascade activation, consistent with non-physiologic intravascular coagulation, in many instances despite little evidence for antibody-mediated complement activation. CONCLUSION We conclude that dysregulated coagulation correlates closely with and probably causes primary failure of pig hearts transgenic for hCRP. These data support efforts to define effective strategies to prevent dysregulated coagulation in pig organ xenografts.
Collapse
Affiliation(s)
- Guosheng Wu
- Baltimore VAMC, University of Maryland, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Pierson RN. Primate T-cell responses to porcine antigens: implications for clinical xenotransplantation. Xenotransplantation 2006; 13:14-8. [PMID: 16497208 DOI: 10.1111/j.1399-3089.2005.00268.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Richard N Pierson
- Department of Surgery, University of Maryland Medical System Staff Surgeon, Baltimore VAMC, Baltimore, MD, USA.
| |
Collapse
|
36
|
Azimzadeh AM, Pfeiffer S, Wu G, Schröder C, Zorn GL, Kelishadi SS, Ozkaynak E, Kehry M, Atkinson JB, Miller GG, Pierson RN. Alloimmunity in primate heart recipients with CD154 blockade: evidence for alternative costimulation mechanisms. Transplantation 2006; 81:255-64. [PMID: 16436970 DOI: 10.1097/01.tp.0000190099.62847.e6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND CD154 mediates key facets of humoral and cellular immunity to alloantigens, and is tolerogenic to influenza antigens in primates. Barriers to CD154-based tolerance induction for primate cardiac allografts have not previously been defined. METHODS Heterotopic cardiac allograft outcomes in cynomolgus monkeys treated with a CD154 inhibitor, IDEC-131 (n=27), were compared to no treatment (n=4) or cyclosporine A (n=6). RESULTS CD154 blockade significantly prolonged median allograft survival, from 6.2 (range 6, 7, n=4) days in untreated controls, to 39 (8,112, n=16) days with intensive monotherapy and 93 (>25, 386; n=3) days with added antithymocyte globulin (ATG), but did not yield tolerance. Alloantibody production was delayed but not prevented by IDEC-131 alone or with ATG, and was exacerbated by infusion of donor bone marrow (n=8). Expression of ICOS was prominent in graft infiltrating lymphocytes, and preceded elaboration of antidonor antibody and vasculopathy. CONCLUSION CD154 monotherapy modulates primate cardiac alloimmunity, but does not readily induce tolerance. Targeting alternative costimulation pathways, including ICOS, may facilitate tolerance induction based on CD154 blockade.
Collapse
Affiliation(s)
- Agnes M Azimzadeh
- Division of Cardiac Surgery, University of Maryland School of Medicine and Baltimore VAMC, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Affiliation(s)
- Reto M Baertschiger
- Surgical Research Unit, Xenotransplantation, Department of Surgery, University Hospital Geneva, Geneva, Switzerland
| | | |
Collapse
|