1
|
Längin M, Bender M, Schmoeckel M, Reichart B. Progress in Orthotopic Pig Heart Transplantation in Nonhuman Primates. Transpl Int 2024; 37:13607. [PMID: 39399753 PMCID: PMC11466817 DOI: 10.3389/ti.2024.13607] [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: 07/31/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024]
Abstract
Xenotransplantation of porcine hearts has become a promising alternative to human allotransplantation, where organ demand still greatly surpasses organ availability. Before entering the clinic, however, feasibility of cardiac xenotransplantation needs to be proven, ideally in the life supporting orthotopic pig-to-nonhuman primate xenotransplantation model. In this review, we shortly outline the last three decades of research and then discuss in detail its most recent advances. These include the genetic modifications of donor pigs to overcome hyperacute rejection and coagulation dysregulation, new organ preservation methods to prevent perioperative xenograft dysfunction, experimental immunosuppressive and immunomodulatory therapies to inhibit the adaptive immune system and systemic inflammation in the recipient, growth control concepts to avoid detrimental overgrowth of the porcine hearts in nonhuman primates, and lastly, the avoidance of porcine cytomegalovirus infections in donor pigs. With these strategies, consistent survival of 6-9 months was achieved in the orthotopic xenotransplantation model, thereby fulfilling the prerequisites for the initiation of a clinical trial.
Collapse
Affiliation(s)
- Matthias Längin
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Martin Bender
- Department of Anesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Michael Schmoeckel
- Department of Cardiac Surgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
2
|
Schmoeckel M, Längin M, Reichart B, Abicht JM, Bender M, Denner J, Marckmann G, Brenner P, Wolf E, Hagl C. [Xenotransplantation of solid organs]. CHIRURGIE (HEIDELBERG, GERMANY) 2024; 95:603-609. [PMID: 38748210 PMCID: PMC11286678 DOI: 10.1007/s00104-024-02093-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 07/30/2024]
Abstract
Transplantation of genetically modified porcine hearts and kidneys could become a solution to the persistent shortage of human organ donors. Progress has been made in genetic engineering of donor pigs, preservation techniques after organ harvesting and immunosuppression using co-stimulation blockade with anti-CD40/CD40L monoclonal antibodies. Progress has also been made in in the development of methods that detect pathogenic porcine viruses and prevent their transmission to the recipient. As normal land breed pig organs continue to grow in the recipient to their original size, different pig breeds (such as Auckland Island pigs) are now used which reach a final size suitable for humans. Alternatively, a knock-out of the growth hormone receptor gene has been established, e.g., in the 10GM genetically modified pigs from Revivicor/United Therapeutics, USA. The first clinical pilot studies including patients suffering from terminal heart failure are expected to start in Germany in about 2 years.
Collapse
Affiliation(s)
- Michael Schmoeckel
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum - Standort Großhadern, Marchioninistr. 15, 81377, München, Deutschland.
| | - Matthias Längin
- Klinik für Anästhesiologie, LMU Klinikum Großhadern, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Bruno Reichart
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, München, Deutschland
| | - Jan-Michael Abicht
- Klinik für Anästhesiologie, LMU Klinikum Großhadern, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Martin Bender
- Klinik für Anästhesiologie, LMU Klinikum Großhadern, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Joachim Denner
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Institut für Virologie, Fachbereich für Veterinärmedizin, FU Berlin, Berlin, Deutschland
| | - Georg Marckmann
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Institut für Ethik, Geschichte und Theorie der Medizin, LMU München, München, Deutschland
| | - Paolo Brenner
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum - Standort Großhadern, Marchioninistr. 15, 81377, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Eckhard Wolf
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Genzentrum und Center for Innovative Medical Models (CIMM), LMU München, München, Deutschland
| | - Christian Hagl
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum - Standort Großhadern, Marchioninistr. 15, 81377, München, Deutschland
- Partner Site München, Deutsches Zentrum für Herz- und Kreislaufforschung e. V. (DZHK), München, Deutschland
| |
Collapse
|
3
|
Peterson L, Yacoub MH, Ayares D, Yamada K, Eisenson D, Griffith BP, Mohiuddin MM, Eyestone W, Venter JC, Smolenski RT, Rothblatt M. Physiological basis for xenotransplantation from genetically modified pigs to humans. Physiol Rev 2024; 104:1409-1459. [PMID: 38517040 PMCID: PMC11390123 DOI: 10.1152/physrev.00041.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
The collective efforts of scientists over multiple decades have led to advancements in molecular and cellular biology-based technologies including genetic engineering and animal cloning that are now being harnessed to enhance the suitability of pig organs for xenotransplantation into humans. Using organs sourced from pigs with multiple gene deletions and human transgene insertions, investigators have overcome formidable immunological and physiological barriers in pig-to-nonhuman primate (NHP) xenotransplantation and achieved prolonged pig xenograft survival. These studies informed the design of Revivicor's (Revivicor Inc, Blacksburg, VA) genetically engineered pigs with 10 genetic modifications (10 GE) (including the inactivation of 4 endogenous porcine genes and insertion of 6 human transgenes), whose hearts and kidneys have now been studied in preclinical human xenotransplantation models with brain-dead recipients. Additionally, the first two clinical cases of pig-to-human heart xenotransplantation were recently performed with hearts from this 10 GE pig at the University of Maryland. Although this review focuses on xenotransplantation of hearts and kidneys, multiple organs, tissues, and cell types from genetically engineered pigs will provide much-needed therapeutic interventions in the future.
Collapse
Affiliation(s)
- Leigh Peterson
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | | | - David Ayares
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - Kazuhiko Yamada
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Daniel Eisenson
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Bartley P Griffith
- University of Maryland Medical Center, Baltimore, Maryland, United States
| | | | - Willard Eyestone
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - J Craig Venter
- J. Craig Venter Institute, Rockville, Maryland, United States
| | | | - Martine Rothblatt
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| |
Collapse
|
4
|
Bender M, Reichart B, Figueiredo C, Burgmann JM, Leuschen M, Wall F, Radan J, Neumann E, Mokelke M, Buttgereit I, Michel S, Ellgass R, Egerer S, Lange A, Baehr A, Kessler B, Kemter E, Klymiuk N, Denner J, Godehardt AW, Tönjes RR, Hagl C, Gebauer M, Binder U, Skerra A, Ayares D, Wolf E, Schmoeckel M, Brenner P, Längin M, Abicht JM. An Approach to Controlling Inflammation and Coagulation in Pig-to-Baboon Cardiac Xenotransplantation. Xenotransplantation 2024; 31:e12877. [PMID: 39077824 DOI: 10.1111/xen.12877] [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: 03/11/2024] [Revised: 06/05/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Inflammatory responses and coagulation disorders are a relevant challenge for successful cardiac xenotransplantation on its way to the clinic. To cope with this, an effective and clinically practicable anti-inflammatory and anti-coagulatory regimen is needed. The inflammatory and coagulatory response can be reduced by genetic engineering of the organ-source pigs. Furthermore, there are several therapeutic strategies to prevent or reduce inflammatory responses and coagulation disorders following xenotransplantation. However, it is still unclear, which combination of drugs should be used in the clinical setting. To elucidate this, we present data from pig-to-baboon orthotopic cardiac xenotransplantation experiments using a combination of several anti-inflammatory drugs. METHODS Genetically modified piglets (GGTA1-KO, hCD46/hTBM transgenic) were used for orthotopic cardiac xenotransplantation into captive-bred baboons (n = 14). All animals received an anti-inflammatory drug therapy including a C1 esterase inhibitor, an IL-6 receptor antagonist, a TNF-α inhibitor, and an IL-1 receptor antagonist. As an additive medication, acetylsalicylic acid and unfractionated heparin were administered. The immunosuppressive regimen was based on CD40/CD40L co-stimulation blockade. During the experiments, leukocyte counts, levels of C-reactive protein (CRP) as well as systemic cytokine and chemokine levels and coagulation parameters were assessed at multiple timepoints. Four animals were excluded from further data analyses due to porcine cytomegalovirus/porcine roseolovirus (PCMV/PRV) infections (n = 2) or technical failures (n = 2). RESULTS Leukocyte counts showed a relevant perioperative decrease, CRP levels an increase. In the postoperative period, leukocyte counts remained consistently within normal ranges, CRP levels showed three further peaks after about 35, 50, and 80 postoperative days. Analyses of cytokines and chemokines revealed different patterns. Some cytokines, like IL-8, increased about 2-fold in the perioperative period, but then decreased to levels comparable to the preoperative values or even lower. Other cytokines, such as IL-12/IL-23, decreased in the perioperative period and stayed at these levels. Besides perioperative decreases, there were no relevant alterations observed in coagulation parameters. In summary, all parameters showed an unremarkable course with regard to inflammatory responses and coagulation disorders following cardiac xenotransplantation and thus showed the effectiveness of our approach. CONCLUSION Our preclinical experience with the anti-inflammatory drug therapy proved that controlling of inflammation and coagulation disorders in xenotransplantation is possible and well-practicable under the condition that transmission of pathogens, especially of PCMV/PRV to the recipient is prevented because PCMV/PRV also induces inflammation and coagulation disorders. Our anti-inflammatory regimen should also be applicable and effective in the clinical setting of cardiac xenotransplantation.
Collapse
Affiliation(s)
- Martin Bender
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Jonathan M Burgmann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Maria Leuschen
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Felicia Wall
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Julia Radan
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Elisabeth Neumann
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Maren Mokelke
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Ines Buttgereit
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Michel
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Reinhard Ellgass
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Egerer
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Andreas Lange
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Andrea Baehr
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Barbara Kessler
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Elisabeth Kemter
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Joachim Denner
- Institute of Virology, Free University Berlin, Berlin, Germany
| | - Antonia W Godehardt
- Division of Haematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Ralf R Tönjes
- Division of Haematology, Cell and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Christian Hagl
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Arne Skerra
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | | | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Michael Schmoeckel
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Paolo Brenner
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Jan-Michael Abicht
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
5
|
Schmoeckel M, Längin M, Reichart B, Abicht JM, Bender M, Michel S, Kamla CE, Denner J, Tönjes RR, Schwinzer R, Marckmann G, Wolf E, Brenner P, Hagl C. Current Status of Cardiac Xenotransplantation: Report of a Workshop of the German Heart Transplant Centers, Martinsried, March 3, 2023. Thorac Cardiovasc Surg 2024; 72:273-284. [PMID: 38154473 PMCID: PMC11147670 DOI: 10.1055/a-2235-8854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023]
Abstract
This report comprises the contents of the presentations and following discussions of a workshop of the German Heart Transplant Centers in Martinsried, Germany on cardiac xenotransplantation. The production and current availability of genetically modified donor pigs, preservation techniques during organ harvesting, and immunosuppressive regimens in the recipient are described. Selection criteria for suitable patients and possible solutions to the problem of overgrowth of the xenotransplant are discussed. Obviously microbiological safety for the recipient and close contacts is essential, and ethical considerations to gain public acceptance for clinical applications are addressed. The first clinical trial will be regulated and supervised by the Paul-Ehrlich-Institute as the National Competent Authority for Germany, and the German Heart Transplant Centers agreed to cooperatively select the first patients for cardiac xenotransplantation.
Collapse
Affiliation(s)
- Michael Schmoeckel
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
| | - Matthias Längin
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Bruno Reichart
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Jan-Michael Abicht
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Martin Bender
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Sebastian Michel
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | | | - Joachim Denner
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Institut für Virologie, Fachbereich für Veterinärmedizin, Freie Universität Berlin, Berlin, Germany
| | - Ralf Reinhard Tönjes
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Paul-Ehrlich-Institut, Langen, Germany
| | - Reinhard Schwinzer
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Georg Marckmann
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Institut für Ethik, Geschichte und Theorie der Medizin, LMU München, Germany
| | - Eckhard Wolf
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Lehrstuhl für Molekulare Tierzucht und Biotechnologie, Genzentrum der LMU München, Germany
| | - Paolo Brenner
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Christian Hagl
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), Partner Site Munich, Germany
| |
Collapse
|
6
|
Schmauch E, Piening B, Mohebnasab M, Xia B, Zhu C, Stern J, Zhang W, Dowdell AK, Kim JI, Andrijevic D, Khalil K, Jaffe IS, Loza BL, Gragert L, Camellato BR, Oliveira MF, O'Brien DP, Chen HM, Weldon E, Gao H, Gandla D, Chang A, Bhatt R, Gao S, Lin X, Reddy KP, Kagermazova L, Habara AH, Widawsky S, Liang FX, Sall J, Loupy A, Heguy A, Taylor SEB, Zhu Y, Michael B, Jiang L, Jian R, Chong AS, Fairchild RL, Linna-Kuosmanen S, Kaikkonen MU, Tatapudi V, Lorber M, Ayares D, Mangiola M, Narula N, Moazami N, Pass H, Herati RS, Griesemer A, Kellis M, Snyder MP, Montgomery RA, Boeke JD, Keating BJ. Integrative multi-omics profiling in human decedents receiving pig heart xenografts. Nat Med 2024; 30:1448-1460. [PMID: 38760586 DOI: 10.1038/s41591-024-02972-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 05/19/2024]
Abstract
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
Collapse
Affiliation(s)
- Eloi Schmauch
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Brian Piening
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Maedeh Mohebnasab
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Jacqueline I Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - David Andrijevic
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Ian S Jaffe
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Bao-Li Loza
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Loren Gragert
- Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | - Han M Chen
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Hui Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Divya Gandla
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Chang
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Riyana Bhatt
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiangping Lin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kriyana P Reddy
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Alawi H Habara
- Department of Biochemistry, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sophie Widawsky
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Joseph Sall
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Alexandre Loupy
- Université Paris Cité, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Adriana Heguy
- Genome Technology Center, NYU Langone Health, New York, NY, USA
| | | | - Yinan Zhu
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Basil Michael
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suvi Linna-Kuosmanen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vasishta Tatapudi
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Navneet Narula
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Nader Moazami
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Harvey Pass
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Ramin S Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | | | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA.
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA.
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Montgomery RA, Griesemer AD, Segev DL, Sommer P. The decedent model: A new paradigm for de-risking high stakes clinical trials like xenotransplantation. Am J Transplant 2024; 24:526-532. [PMID: 38341026 DOI: 10.1016/j.ajt.2024.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
The first 2 living recipients of pig hearts died unexpectedly within 2 months, despite both recipients receiving what over 30 years of nonhuman primate (NHP) research would suggest were the optimal gene edits and immunosuppression to ensure success. These results prompt us to question how faithfully data from the NHP model translate into human outcomes. Before attempting any further heart xenotransplants in living humans, it is highly advisable to gain a more comprehensive understanding of why the promising preclinical NHP data did not accurately predict outcomes in humans. It is also unlikely that additional NHP data will provide more information that would de-risk a xenoheart clinical trial because these cases were based on the best practices from the most successful NHP results to date. Although imperfect, the decedent model offers a complementary avenue to determine appropriate treatment regimens to control the human immune response to xenografts and better understand the biologic differences between humans and NHP that could lead to such starkly contrasting outcomes. Herein, we explore the potential benefits and drawbacks of the decedent model and contrast it to the advantages and disadvantages of the extensive body of data generated in the NHP xenoheart transplantation model.
Collapse
Affiliation(s)
| | | | | | - Philip Sommer
- NYU Langone Transplant Institute, New York, NY, USA; NYU Department of Anesthesiology, Perioperative Care and Pain Medicine, New York, NY, USA
| |
Collapse
|
8
|
Bender M, Panelli A, Reichart B, Radan J, Mokelke M, Neumann E, Buttgereit I, Michel S, Bauer A, Fresch AK, Mayr T, Werner F, Egerer S, Bähr A, Kessler B, Klymiuk N, Ayares D, Wolf E, Hagl C, Brenner P, Längin M, Abicht J. Hemodynamics in pig-to-baboon heterotopic thoracic cardiac xenotransplantation: Recovery from perioperative cardiac xenograft dysfunction and impairment by cardiac overgrowth. Xenotransplantation 2024; 31:e12841. [PMID: 38864375 PMCID: PMC11167678 DOI: 10.1111/xen.12841] [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: 04/04/2023] [Revised: 09/04/2023] [Accepted: 12/19/2023] [Indexed: 06/13/2024]
Abstract
INTRODUCTION Orthotopic cardiac xenotransplantation has seen notable improvement, leading to the first compassionate use in 2022. However, it remains challenging to define the clinical application of cardiac xenotransplantation, including the back-up strategy in case of xenograft failure. In this regard, the heterotopic thoracic technique could be an alternative to the orthotopic procedure. We present hemodynamic data of heterotopic thoracic pig-to-baboon transplantation experiments, focusing on perioperative xenograft dysfunction and xenograft overgrowth. METHODS We used 17 genetically modified piglets as donors for heterotopic thoracic xenogeneic cardiac transplantation into captive-bred baboons. In all animals, pressure probes were implanted in the graft's left ventricle and the recipient's ascending aorta and hemodynamic data (graft pressure, aortic pressure and recipient's heart rate) were recorded continuously. RESULTS Aortic pressures and heart rates of the recipients' hearts were postoperatively stable in all experiments. After reperfusion, three grafts presented with low left ventricular pressure indicating perioperative cardiac dysfunction (PCXD). These animals recovered from PCXD within 48 h under support of the recipient's heart and there was no difference in survival compared to the other 14 ones. After 48 h, graft pressure increased up to 200 mmHg in all 17 animals with two different time-patterns. This led to a progressive gradient between graft and aortic pressure. With increasing gradient, the grafts stopped contributing to cardiac output. Grafts showed a marked weight increase from implantation to explantation. CONCLUSION The heterotopic thoracic cardiac xenotransplantation technique is a possible method to overcome PCXD in early clinical trials and an experimental tool to get a better understanding of PCXD. The peculiar hemodynamic situation of increasing graft pressure but missing graft's output indicates outflow tract obstruction due to cardiac overgrowth. The heterotopic thoracic technique should be successful when using current strategies of immunosuppression, organ preservation and donor pigs with smaller body and organ size.
Collapse
Affiliation(s)
- Martin Bender
- Department of AnaesthesiologyUniversity Hospital, LMU MunichMunichGermany
| | - Alessandro Panelli
- Department of Anesthesiology and Operative Intensive Care Medicine (CVK, CCM)Charité – Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Bruno Reichart
- Transregional Collaborative Research Center 127Walter Brendel Centre of Experimental MedicineLMU MunichMunichGermany
| | - Julia Radan
- Transregional Collaborative Research Center 127Walter Brendel Centre of Experimental MedicineLMU MunichMunichGermany
| | - Maren Mokelke
- Transregional Collaborative Research Center 127Walter Brendel Centre of Experimental MedicineLMU MunichMunichGermany
| | - Elisabeth Neumann
- Transregional Collaborative Research Center 127Walter Brendel Centre of Experimental MedicineLMU MunichMunichGermany
| | - Ines Buttgereit
- Department of AnaesthesiologyUniversity Hospital, LMU MunichMunichGermany
| | - Sebastian Michel
- Department of Cardiac SurgeryUniversity HospitalLMU MunichMunichGermany
| | - Andreas Bauer
- Department of AnaesthesiologyUniversity Hospital, LMU MunichMunichGermany
| | - Ann Kathrin Fresch
- Transregional Collaborative Research Center 127Walter Brendel Centre of Experimental MedicineLMU MunichMunichGermany
| | - Tanja Mayr
- Department of AnaesthesiologyUniversity Hospital, LMU MunichMunichGermany
| | - Fabian Werner
- Transregional Collaborative Research Center 127Walter Brendel Centre of Experimental MedicineLMU MunichMunichGermany
| | - Stefanie Egerer
- Gene Center and Department of Veterinary SciencesInstitute of Molecular Animal Breeding and BiotechnologyLMU MunichMunichGermany
| | - Andrea Bähr
- Gene Center and Department of Veterinary SciencesInstitute of Molecular Animal Breeding and BiotechnologyLMU MunichMunichGermany
| | - Barbara Kessler
- Gene Center and Department of Veterinary SciencesInstitute of Molecular Animal Breeding and BiotechnologyLMU MunichMunichGermany
| | - Nikolai Klymiuk
- Gene Center and Department of Veterinary SciencesInstitute of Molecular Animal Breeding and BiotechnologyLMU MunichMunichGermany
| | | | - Eckhard Wolf
- Gene Center and Department of Veterinary SciencesInstitute of Molecular Animal Breeding and BiotechnologyLMU MunichMunichGermany
- Center for Innovative Medical Models (CiMM)LMU MunichMunichGermany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU)LMU MunichMunichGermany
| | - Christian Hagl
- Department of Cardiac SurgeryUniversity HospitalLMU MunichMunichGermany
| | - Paolo Brenner
- Department of Cardiac SurgeryUniversity HospitalLMU MunichMunichGermany
| | - Matthias Längin
- Department of AnaesthesiologyUniversity Hospital, LMU MunichMunichGermany
| | - Jan‐Michael Abicht
- Department of AnaesthesiologyUniversity Hospital, LMU MunichMunichGermany
| |
Collapse
|
9
|
Burdorf L, Gao Z, Riner A, Sievert E, Harris DG, Kuravi KV, Morrill BH, Habibabady Z, Rybak E, Dahi S, Zhang T, Schwartz E, Kang E, Cheng X, Esmon CT, Phelps CJ, Ayares DL, Pierson RN, Azimzadeh AM. Expression of human thrombomodulin by GalTKO.hCD46 pigs modulates coagulation cascade activation by endothelial cells and during ex vivo lung perfusion with human blood. Xenotransplantation 2023; 30:e12828. [PMID: 37767640 PMCID: PMC10840969 DOI: 10.1111/xen.12828] [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: 12/08/2020] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Thrombomodulin is important for the production of activated protein C (APC), a molecule with significant regulatory roles in coagulation and inflammation. To address known molecular incompatibilities between pig thrombomodulin and human thrombin that affect the conversion of protein C into APC, GalTKO.hCD46 pigs have been genetically modified to express human thrombomodulin (hTBM). The aim of this study was to evaluate the impact of transgenic hTBM expression on the coagulation dysregulation that is observed in association with lung xenograft injury in an established lung perfusion model, with and without additional blockade of nonphysiologic interactions between pig vWF and human GPIb axis. Expression of hTBM was variable between pigs at the transcriptional and protein level. hTBM increased the activation of human protein C and inhibited thrombosis in an in vitro flow perfusion assay, confirming that the expressed protein was functional. Decreased platelet activation was observed during ex vivo perfusion of GalTKO.hCD46 lungs expressing hTBM and, in conjunction with transgenic hTBM, blockade of the platelet GPIb receptor further inhibited platelets and increased survival time. Altogether, our data indicate that expression of transgenic hTBM partially addresses coagulation pathway dysregulation associated with pig lung xenograft injury and, in combination with vWF-GP1b-directed strategies, is a promising approach to improve the outcomes of lung xenotransplantation.
Collapse
Affiliation(s)
- Lars Burdorf
- Center for Transplantation Sciences, Department of Surgery,
Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Zhuo Gao
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing
Medical University, Nanjing, China, USA
| | - Andrea Riner
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Evelyn Sievert
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Donald G. Harris
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | | | | | - Zahra Habibabady
- Center for Transplantation Sciences, Department of Surgery,
Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Elana Rybak
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Siamak Dahi
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Tianshu Zhang
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Evan Schwartz
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Elizabeth Kang
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Xiangfei Cheng
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Charles T. Esmon
- Cardiovascular Biology Research Program, Oklahoma Medical
Research Foundation, Department of Pathology, University of Oklahoma Health Sciences
Center, Oklahoma City, OK, USA
| | | | | | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery,
Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| | - Agnes M. Azimzadeh
- Center for Transplantation Sciences, Department of Surgery,
Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, University of Maryland School of
Medicine, and VA Maryland Health Care System, Baltimore, MD, USA
| |
Collapse
|
10
|
Moazami N, Stern JM, Khalil K, Kim JI, Narula N, Mangiola M, Weldon EP, Kagermazova L, James L, Lawson N, Piper GL, Sommer PM, Reyentovich A, Bamira D, Saraon T, Kadosh BS, DiVita M, Goldberg RI, Hussain ST, Chan J, Ngai J, Jan T, Ali NM, Tatapudi VS, Segev DL, Bisen S, Jaffe IS, Piegari B, Kowalski H, Kokkinaki M, Monahan J, Sorrells L, Burdorf L, Boeke JD, Pass H, Goparaju C, Keating B, Ayares D, Lorber M, Griesemer A, Mehta SA, Smith DE, Montgomery RA. Pig-to-human heart xenotransplantation in two recently deceased human recipients. Nat Med 2023; 29:1989-1997. [PMID: 37488288 DOI: 10.1038/s41591-023-02471-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/26/2023] [Indexed: 07/26/2023]
Abstract
Genetically modified xenografts are one of the most promising solutions to the discrepancy between the numbers of available human organs for transplantation and potential recipients. To date, a porcine heart has been implanted into only one human recipient. Here, using 10-gene-edited pigs, we transplanted porcine hearts into two brain-dead human recipients and monitored xenograft function, hemodynamics and systemic responses over the course of 66 hours. Although both xenografts demonstrated excellent cardiac function immediately after transplantation and continued to function for the duration of the study, cardiac function declined postoperatively in one case, attributed to a size mismatch between the donor pig and the recipient. For both hearts, we confirmed transgene expression and found no evidence of cellular or antibody-mediated rejection, as assessed using histology, flow cytometry and a cytotoxic crossmatch assay. Moreover, we found no evidence of zoonotic transmission from the donor pigs to the human recipients. While substantial additional work will be needed to advance this technology to human trials, these results indicate that pig-to-human heart xenotransplantation can be performed successfully without hyperacute rejection or zoonosis.
Collapse
Affiliation(s)
- Nader Moazami
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA.
| | - Jeffrey M Stern
- New York University Langone Transplant Institute, New York, NY, USA
| | - Karen Khalil
- New York University Langone Transplant Institute, New York, NY, USA
| | - Jacqueline I Kim
- New York University Langone Transplant Institute, New York, NY, USA
| | - Navneet Narula
- Department of Pathology, New York University Langone Health, New York, NY, USA
| | - Massimo Mangiola
- New York University Langone Transplant Institute, New York, NY, USA
| | - Elaina P Weldon
- New York University Langone Transplant Institute, New York, NY, USA
| | - Larisa Kagermazova
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Les James
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | - Nikki Lawson
- New York University Langone Transplant Institute, New York, NY, USA
| | - Greta L Piper
- Department of Surgery, New York University Langone Health, New York, NY, USA
| | - Philip M Sommer
- Department of Anesthesiology, New York University Langone Health, New York, NY, USA
| | - Alex Reyentovich
- Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Daniel Bamira
- Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Tajinderpal Saraon
- Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Bernard S Kadosh
- Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Michael DiVita
- Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Randal I Goldberg
- Division of Cardiology, New York University Langone Health, New York, NY, USA
| | - Syed T Hussain
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | - Justin Chan
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | - Jennie Ngai
- Department of Anesthesiology, New York University Langone Health, New York, NY, USA
| | - Thomas Jan
- Department of Anesthesiology, New York University Langone Health, New York, NY, USA
| | - Nicole M Ali
- New York University Langone Transplant Institute, New York, NY, USA
| | | | - Dorry L Segev
- Department of Surgery, New York University Langone Health, New York, NY, USA
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, USA
| | - Shivani Bisen
- New York University Grossman School of Medicine, New York University, New York, NY, USA
| | - Ian S Jaffe
- New York University Grossman School of Medicine, New York University, New York, NY, USA
| | - Benjamin Piegari
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Haley Kowalski
- New York University Grossman School of Medicine, New York University, New York, NY, USA
| | | | | | | | | | - Jef D Boeke
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, USA
| | - Harvey Pass
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | - Chandra Goparaju
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | - Brendan Keating
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Marc Lorber
- United Therapeutics Corporation, Silver Spring, MD, USA
| | - Adam Griesemer
- New York University Langone Transplant Institute, New York, NY, USA
| | - Sapna A Mehta
- New York University Langone Transplant Institute, New York, NY, USA
| | - Deane E Smith
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | | |
Collapse
|
11
|
Tseng HT, Lin YW, Huang CY, Shih CM, Tsai YT, Liu CW, Tsai CS, Lin FY. Animal Models for Heart Transplantation Focusing on the Pathological Conditions. Biomedicines 2023; 11:biomedicines11051414. [PMID: 37239085 DOI: 10.3390/biomedicines11051414] [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: 03/30/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiac transplant recipients face many complications due to transplant rejection. Scientists must conduct animal experiments to study disease onset mechanisms and develop countermeasures. Therefore, many animal models have been developed for research topics including immunopathology of graft rejection, immunosuppressive therapies, anastomotic techniques, and graft preservation techniques. Small experimental animals include rodents, rabbits, and guinea pigs. They have a high metabolic rate, high reproductive rate, small size for easy handling, and low cost. Additionally, they have genetically modified strains for pathological mechanisms research; however, there is a lacuna, as these research results rarely translate directly to clinical applications. Large animals, including canines, pigs, and non-human primates, have anatomical structures and physiological states that are similar to those of humans; therefore, they are often used to validate the results obtained from small animal studies and directly speculate on the feasibility of applying these results in clinical practice. Before 2023, PubMed Central® at the United States National Institute of Health's National Library of Medicine was used for literature searches on the animal models for heart transplantation focusing on the pathological conditions. Unpublished reports and abstracts from conferences were excluded from this review article. We discussed the applications of small- and large-animal models in heart transplantation-related studies. This review article aimed to provide researchers with a complete understanding of animal models for heart transplantation by focusing on the pathological conditions created by each model.
Collapse
Affiliation(s)
- Horng-Ta Tseng
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Wen Lin
- Institute of Oral Biology, National Yang Ming Chiao Tung University (Yangming Campus), Taipei 112304, Taiwan
| | - Chun-Yao Huang
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Ming Shih
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, Defense Medical Center, Taipei 11490, Taiwan
| | - Chen-Wei Liu
- Department of Basic Medical Science, College of Medicine, University of Arizona, Phoenix, AZ 85721, USA
| | - Chien-Sung Tsai
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiovascular Surgery, Tri-Service General Hospital, Defense Medical Center, Taipei 11490, Taiwan
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Feng-Yen Lin
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
12
|
Rivera NT, Baran DA. Expanding heart transplantation in 2022 and beyond. Curr Opin Cardiol 2023; 38:130-135. [PMID: 36598449 DOI: 10.1097/hco.0000000000001023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Despite advances in the technology of mechanical circulatory support, the need for heart transplantation continues to grow. The longevity of heart transplants continues to be superior to mechanical solutions, though the short-term differences are shrinking. In this review, we cover three timely developments and summarize the recent literature. RECENT FINDINGS After stagnant rates of heart transplant activity for some years, recently, transplant volume has increased. The developments that have ignited interest have been the use of hepatitis C infected donors, which can now be safely transplanted with the advent of curative oral regimens, and the worldwide use of donors following withdrawal of life support as opposed to traditional brain death donors. In addition, the recent experience of human cardiac xenotransplantation has been very exciting, and though it is not of clinical utility yet, it holds the promise for a virtually unlimited supply of organs at some time in the future. SUMMARY Much work remains to be done, but together, all three of these developments are exciting and important to be aware of in the future. Each will contribute to additional donors for human heart transplantation and hopefully will alleviate suffering and death on the waiting list.
Collapse
|
13
|
Reichart B, Cooper DKC, Längin M, Tönjes RR, Pierson RN, Wolf E. Cardiac xenotransplantation: from concept to clinic. Cardiovasc Res 2023; 118:3499-3516. [PMID: 36461918 PMCID: PMC9897693 DOI: 10.1093/cvr/cvac180] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 12/05/2022] Open
Abstract
For many patients with terminal/advanced cardiac failure, heart transplantation is the most effective, durable treatment option, and offers the best prospects for a high quality of life. The number of potentially life-saving donated human organs is far fewer than the population who could benefit from a new heart, resulting in increasing numbers of patients awaiting replacement of their failing heart, high waitlist mortality, and frequent reliance on interim mechanical support for many of those deemed among the best candidates but who are deteriorating as they wait. Currently, mechanical assist devices supporting left ventricular or biventricular heart function are the only alternative to heart transplant that is in clinical use. Unfortunately, the complication rate with mechanical assistance remains high despite advances in device design and patient selection and management, and the quality of life of the patients even with good outcomes is only moderately improved. Cardiac xenotransplantation from genetically multi-modified (GM) organ-source pigs is an emerging new option as demonstrated by the consistent long-term success of heterotopic (non-life-supporting) abdominal and life-supporting orthotopic porcine heart transplantation in baboons, and by a recent 'compassionate use' transplant of the heart from a GM pig with 10 modifications into a terminally ill patient who survived for 2 months. In this review, we discuss pig heart xenotransplantation as a concept, including pathobiological aspects related to immune rejection, coagulation dysregulation, and detrimental overgrowth of the heart, as well as GM strategies in pigs to prevent or minimize these problems. Additional topics discussed include relevant results of heterotopic and orthotopic heart transplantation experiments in the pig-to-baboon model, microbiological and virologic safety concepts, and efficacy requirements for initiating formal clinical trials. An adequate regulatory and ethical framework as well as stringent criteria for the selection of patients will be critical for the safe clinical development of cardiac xenotransplantation, which we expect will be clinically tested during the next few years.
Collapse
Affiliation(s)
- Bruno Reichart
- Walter Brendel Centre for Experimental Medicine, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - David K C Cooper
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Ralf R Tönjes
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, Langen 63225, Germany
| | - Richard N Pierson
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Eckhard Wolf
- Gene Centre and Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Munich 81377, Germany
| |
Collapse
|
14
|
Mohiuddin MM, Singh AK, Goerlich CE. Preclinical rationale and current pathways to support the first human clinical trials in cardiac xenotransplantation. Hum Immunol 2023; 84:34-42. [PMID: 35851182 PMCID: PMC10154071 DOI: 10.1016/j.humimm.2022.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/29/2022] [Accepted: 07/07/2022] [Indexed: 01/05/2023]
Abstract
Recent initiation of the first FDA-approved cardiac xenotransplantation suggests xenotransplantation could soon become a therapeutic option for patients unable to undergo allotransplantation. Until xenotransplantation is widely applied in clinical practice, consideration of benefit versus risk and approaches to management of clinical xenografts will based at least in part on observations made in experimental xenotransplantation in non-human primates. Indeed, the decision to proceed with clinical trials reflects significant progress in last few years in experimental solid organ and cellular xenotransplantation. Our laboratory at the NIH and now at University of Maryland contributed to this progress, with heterotopic cardiac xenografts surviving more than two years and life-supporting cardiac xenografts survival up to 9 months. Here we describe our contributions to the understanding of the mechanism of cardiac xenograft rejection and development of methods to overcome past hurdles, and finally we share our opinion on the remaining barriers to clinical translation. We also discuss how the first in human xenotransplants might be performed, recipients managed, and graft function monitored.
Collapse
|
15
|
Garry DJ, Weiner JI, Greising SM, Garry MG, Sachs DH. Mechanisms and strategies to promote cardiac xenotransplantation. J Mol Cell Cardiol 2022; 172:109-119. [PMID: 36030840 DOI: 10.1016/j.yjmcc.2022.07.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/21/2022] [Accepted: 07/31/2022] [Indexed: 12/14/2022]
Abstract
End stage heart failure is a terminal disease, and the only curative therapy is orthotopic heart transplantation. Due to limited organ availability, alternative strategies have received intense interest for treatment of patients with advanced heart failure. Recent studies using gene-edited porcine organs suggest that cardiac xenotransplantation may provide a future source of organs. In this review, we highlight the historical milestones for cardiac xenotransplantation and the gene editing strategies designed to overcome immunological barriers, which have culminated in a recent cardiac pig-to-human xenotransplant. We also discuss recent results of studies on the engineering of human-porcine chimeric organs that may provide an alternative and complementary strategy to overcome some of the major immunological barriers to producing a new source of transplantable organs.
Collapse
Affiliation(s)
- Daniel J Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, United States of America; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, United States of America; NorthStar Genomics, Eagan, MN, United States of America.
| | - Joshua I Weiner
- Departments of Surgery, Columbia Center for Translational Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States of America
| | - Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Mary G Garry
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, United States of America; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, United States of America; NorthStar Genomics, Eagan, MN, United States of America
| | - David H Sachs
- Departments of Surgery, Columbia Center for Translational Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States of America; Department of Surgery, Massachusetts General Hospital, Boston, MA, United States of America
| |
Collapse
|
16
|
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
|
17
|
Raza SS, Hara H, Cleveland DC, Cooper DKC. The potential of genetically engineered pig heart transplantation in infants with complex congenital heart disease. Pediatr Transplant 2022; 26:e14260. [PMID: 35233893 PMCID: PMC10124767 DOI: 10.1111/petr.14260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022]
Abstract
Despite advances in surgical and medical techniques, complex congenital heart disease in neonates and infants continues to be associated with significant mortality and morbidity. More than 500 infants in the USA are placed on the cardiac transplantation wait-list annually. However, there remains a critical shortage of deceased human donor organs for transplantation with a median wait-time of 4 months. Hence, infant mortality on the heart transplant wait-list in the USA is higher than for any other solid organ transplant group. Orthotopic transplantation of a pig heart as a bridge to allotransplantation might offer the best prospect of long-term survival of these patients. In recent years, there have been several advances in genetic engineering of pigs to mitigate the vigorous antibody-mediated rejection of a pig heart transplanted into a nonhuman primate. In this review, we briefly highlight (i) the history of clinical heart xenotransplantation, (ii) current advances and techniques of genetically engineering pigs, (iii) the current status of pig orthotopic cardiac graft survival in nonhuman primates, and (iv) progress toward pursuing clinical trials of cardiac xenotransplantation. Ultimately, we argue that pig heart xenotransplantation should initially be used as a bridge to cardiac allotransplantation in neonates and infants.
Collapse
Affiliation(s)
- Syed Sikandar Raza
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David C Cleveland
- Department of Cardiothoracic Surgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
18
|
Litovsky SH, Foote JB, Jagdale A, Walcott G, Iwase H, Bikhet MH, Yamamoto T, Hansen-Estruch C, Ezzelarab MB, Ayares D, Carlo WF, Rhodes LA, Crawford JH, Borasino S, Dabal RJ, Padilla LA, Hara H, Cooper DK, Cleveland DC. Cardiac and Pulmonary Histopathology in Baboons Following Genetically-Engineered Pig Orthotopic Heart Transplantation. Ann Transplant 2022; 27:e935338. [PMID: 35789146 PMCID: PMC9270855 DOI: 10.12659/aot.935338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/20/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Although improving, survival after pig orthotopic heart transplantation (OHTx) in baboons has been mixed and largely poor. The causes for the high incidence of early failure remain uncertain. MATERIAL AND METHODS We have carried out pig OHTx in 4 baboons. Two died or were euthanized within hours, and 2 survived for 3 and 8 months, respectively. There was evidence of a significant 'cytokine storm' in the immediate post-OHTx period with the elevations in IL-6 correlating closely with the final outcome. RESULTS All 4 baboons demonstrated features suggestive of respiratory dysfunction, including increased airway resistance, hypoxia, and tachypnea. Histopathological observations of pulmonary infiltration by neutrophils and, notably, eosinophils within vessels and in the perivascular and peribronchiolar space, with minimal cardiac pathology, suggested a role for early lung acute inflammation. In one, features suggestive of transfusion-related acute lung injury were present. The 2 longer-term survivors died of (i) a cardiac dysrhythmia with cellular infiltration around the conducting tissue (at 3 months), and (ii) mixed cellular and antibody-mediated rejection (at 8 months). CONCLUSIONS These initial findings indicate a potential role of acute lung injury early after OHTx. If this response can be prevented, increased survival may result, providing an opportunity to evaluate the factors affecting long-term survival.
Collapse
Affiliation(s)
- Silvio H. Litovsky
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeremy B. Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Abhijit Jagdale
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory Walcott
- Division of Cardiovascular Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohamed H. Bikhet
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Takayuki Yamamoto
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christophe Hansen-Estruch
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohamed B. Ezzelarab
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Waldemar F. Carlo
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leslie A. Rhodes
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jack H. Crawford
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Santiago Borasino
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert J. Dabal
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Luz A. Padilla
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K.C. Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David C. Cleveland
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
19
|
Boulet J, Cunningham JW, Mehra MR. Cardiac Xenotransplantation. JACC Basic Transl Sci 2022; 7:716-729. [PMID: 35958689 PMCID: PMC9357575 DOI: 10.1016/j.jacbts.2022.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 10/27/2022]
|
20
|
Miura S, Habibabady ZA, Pollok F, Connolly M, Pratts S, Dandro A, Sorrells L, Karavi K, Phelps C, Eyestone W, Ayares D, Burdorf L, Azimzadeh A, Pierson RN. Effects of human TFPI and CD47 expression and selectin and integrin inhibition during GalTKO.hCD46 pig lung perfusion with human blood. Xenotransplantation 2022; 29:e12725. [PMID: 35234315 PMCID: PMC10207735 DOI: 10.1111/xen.12725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/30/2021] [Accepted: 12/17/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Loss of barrier function when GalTKO.hCD46 porcine lungs are perfused with human blood is associated with coagulation pathway dysregulation, innate immune system activation, and rapid sequestration of human formed blood elements. Here, we evaluate whether genetic expression of human tissue factor pathway inhibitor (hTFPI) and human CD47 (hCD47), alone or with combined selectin and integrin adhesion pathway inhibitors, delays GalTKO.hCD46 porcine lung injury or modulates neutrophil and platelet sequestration. METHODS In a well-established paired ex vivo lung perfusion model, GalTKO.hCD46.hTFPI.hCD47 transgenic porcine lungs (hTFPI.hCD47, n = 7) were compared to GalTKO.hCD46 lungs (reference, n = 5). All lung donor pigs were treated with a thromboxane synthase inhibitor, anti-histamine, and anti-GPIb integrin-blocking Fab, and were pre-treated with Desmopressin. In both genotypes, one lung of each pair was additionally treated with PSGL-1 and GMI-1271 (P- and E-selectin) and IB4 (CD11b/18 integrin) adhesion inhibitors (n = 6 hTFPI.hCD47, n = 3 reference). RESULTS All except for two reference lungs did not fail within 480 min when experiments were electively terminated. Selectin and integrin adhesion inhibitors moderately attenuated initial pulmonary vascular resistance (PVR) elevation in hTFPI.hCD47 lungs. Neutrophil sequestration was significantly delayed during the early time points following reperfusion and terminal platelet activation was attenuated in association with lungs expressing hTFPI.hCD47, but additional adhesion pathway inhibitors did not show further effects with either lung genotype. CONCLUSION Expression of hTFPI.hCD47 on porcine lung may be useful as part of an integrated strategy to prevent neutrophil adhesion and platelet activation that are associated with xenograft injury. Additionally, targeting canonical selectin and integrin adhesion pathways reduced PVR elevation associated with hTFPI.hCD47 expression, but did not significantly attenuate neutrophil or platelet sequestration. We conclude that other adhesive mechanisms mediate the residual sequestration of human formed blood elements to pig endothelium that occurs even in the context of the multiple genetic modifications and drug treatments tested here.
Collapse
Affiliation(s)
- Shuhei Miura
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Cardiovascular Surgery, Teine Keijinkai Hospital, Sapporo, Japan
| | - Zahra A. Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Franziska Pollok
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Margaret Connolly
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shannon Pratts
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | - Lars Burdorf
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Agnes Azimzadeh
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Song M, Tang Z, Liu Y, Xie X, Qi X, Wu Q, Jiang Z, Wu Z, Qian T. Yak Pericardium as an Alternative Biomaterial for Transcatheter Heart Valves. Front Bioeng Biotechnol 2021; 9:766991. [PMID: 34820366 PMCID: PMC8607193 DOI: 10.3389/fbioe.2021.766991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
Transcatheter aortic valve implantation (TAVI) has received much attention and development in the past decade due to its lower risk of complication and infections compared to a traditional open thoracotomy. However, the current commercial transcatheter heart valve does not fully meet clinical needs; therefore, new biological materials must be found in order to meet these requirements. We have discovered a new type of biological material, the yak pericardium. This current research studied its extracellular matrix structure, composition, mechanical properties, and amino acid content. Folding experiment was carried out to analyze the structure and mechanics after folding. We also conducted a subcutaneous embedding experiment to analyze the inflammatory response and calcification after implantation. Australian bovine pericardium, local bovine pericardium, and porcine pericardium were used as controls. The overall structure of the yak pericardium is flat, the collagen runs regularly, it has superior mechanical properties, and the average thickness is significantly lower than that of the Australian bovine and the local bovine pericardium control groups. The yak pericardium has a higher content of elastic fibers, showing that it has a better compression resistance effect during the folding experiment as well as having less expression of transplantation-related antigens. We conducted in vivo experiments and found that the yak pericardium has less inflammation and a lower degree of calcification. In summary, the yak pericardium, which is thin and strong, has lower immunogenicity and outstanding anti-calcification effects may be an excellent candidate valve leaflet material for TAVI.
Collapse
Affiliation(s)
- Mingzhe Song
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Zhenjie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Yuhong Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Xinlong Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Xiaoke Qi
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Qiying Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Zhenlin Jiang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Zhongshi Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Tao Qian
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| |
Collapse
|
22
|
Reichart B, Längin M, Denner J, Schwinzer R, Cowan PJ, Wolf E. Pathways to Clinical Cardiac Xenotransplantation. Transplantation 2021; 105:1930-1943. [PMID: 33350675 DOI: 10.1097/tp.0000000000003588] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart transplantation is the only long-lasting lifesaving option for patients with terminal cardiac failure. The number of available human organs is however far below the actual need, resulting in substantial mortality of patients while waiting for a human heart. Mechanical assist devices are used to support cardiac function but are associated with a high risk of severe complications and poor quality of life for the patients. Consistent success in orthotopic transplantation of genetically modified pig hearts into baboons indicates that cardiac xenotransplantation may become a clinically applicable option for heart failure patients who cannot get a human heart transplant. In this overview, we project potential paths to clinical cardiac xenotransplantation, including the choice of genetically modified source pigs; associated requirements of microbiological, including virological, safety; optimized matching of source pig and recipient; and specific treatments of the donor heart after explantation and of the recipients. Moreover, selection of patients and the regulatory framework will be discussed.
Collapse
Affiliation(s)
- Bruno Reichart
- Walter Brendel Center for Experimental Medicine, LMU Munich, Munich, Germany
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Joachim Denner
- Institute of Virology, Free University Berlin, Berlin, Germany
| | - Reinhard Schwinzer
- Department of General-, Visceral-, and Transplantation Surgery, Transplant Laboratory, Hannover Medical School, Hannover, Germany
| | - Peter J Cowan
- Immunology Research Centre, St. Vincent's Hospital Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, VIC, Australia
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
- Department of Veterinary Sciences, and Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| |
Collapse
|
23
|
Goerlich CE, Griffith B, Singh AK, Abdullah M, Singireddy S, Kolesnik I, Lewis B, Sentz F, Tatarov I, Hershfeld A, Zhang T, Strauss E, Odonkor P, Williams B, Tabatabai A, Bhutta A, Ayares D, Kaczorowski DJ, Mohiuddin MM. Blood Cardioplegia Induction, Perfusion Storage and Graft Dysfunction in Cardiac Xenotransplantation. Front Immunol 2021; 12:667093. [PMID: 34177906 PMCID: PMC8220198 DOI: 10.3389/fimmu.2021.667093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/18/2021] [Indexed: 01/05/2023] Open
Abstract
Background Perioperative cardiac xenograft dysfunction (PCXD) describes a rapidly developing loss of cardiac function after xenotransplantation. PCXD occurs despite genetic modifications to increase compatibility of the heart. We report on the incidence of PCXD using static preservation in ice slush following crystalloid or blood-based cardioplegia versus continuous cold perfusion with XVIVO© heart solution (XHS) based cardioplegia. Methods Baboons were weight matched to genetically engineered swine heart donors. Cardioplegia volume was 30 cc/kg by donor weight, with del Nido cardioplegia and the addition of 25% by volume of donor whole blood. Continuous perfusion was performed using an XVIVO © Perfusion system with XHS to which baboon RBCs were added. Results PCXD was observed in 5/8 that were preserved with crystalloid cardioplegia followed by traditional cold, static storage on ice. By comparison, when blood cardioplegia was used followed by cold, static storage, PCXD occurred in 1/3 hearts and only in 1/5 hearts that were induced with XHS blood cardioplegia followed by continuous perfusion. Survival averaged 17 hours in those with traditional preservation and storage, followed by 11.47 days and 15.03 days using blood cardioplegia and XHS+continuous preservation, respectively. Traditional preservation resulted in more inotropic support and higher average peak serum lactate 14.3±1.7 mmol/L compared to blood cardioplegia 3.6±3.0 mmol/L and continuous perfusion 3.5±1.5 mmol/L. Conclusion Blood cardioplegia induction, alone or followed by XHS perfusion storage, reduced the incidence of PCXD and improved graft function and survival, relative to traditional crystalloid cardioplegia-slush storage alone.
Collapse
Affiliation(s)
- Corbin E Goerlich
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Bartley Griffith
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Avneesh K Singh
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mohamed Abdullah
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Cardiothoracic Surgery, Cairo University, Cairo, Egypt
| | - Shreya Singireddy
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina Kolesnik
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Billeta Lewis
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Faith Sentz
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ivan Tatarov
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alena Hershfeld
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tianshu Zhang
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Erik Strauss
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Patrick Odonkor
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Brittney Williams
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ali Tabatabai
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Adnan Bhutta
- Department of Pediatrics, The University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - David J Kaczorowski
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| | - Muhammad M Mohiuddin
- Department of Surgery, The University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
24
|
Tomasi R, Tariq M, Hübner M, Strauss G, Längin M, Zeuzem-Lampert C, Vandewiele S, Kreth S, Abicht JM. T-Cell Response in a Cardiac Xenotransplant Model. EXP CLIN TRANSPLANT 2021; 19:708-716. [PMID: 34085920 DOI: 10.6002/ect.2020.0359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Despite the advances in preclinical cardiac xenotransplantation, the immune reactions caused by species differences are not fully understood. Hyperacute rejection can now be avoided using genetically engineered donor organs, but cellmediated rejection by the adaptive immune response has not been addressed successfully. Here we investigated the initial human pan-T-cell reaction using a pig-human blood working heart model. MATERIALS AND METHODS Porcine wild-type hearts (n = 7) were perfused with human blood in a biventricular working heart system for 3 hours. As control, blood from the same human donors was circulated without a pig heart. Pan-T cells were selectively extracted from blood taken before and at the end of the perfusion cycle. The relative mRNA expression of selected target genes (real-time quantitative polymerase chain reaction) and the expression of microRNAs were determined. RESULTS After xenogeneic organ perfusion, there was a moderate upregulation of several CD4+ marker cytokines (interleukin 2, interleukin 4, interferon γ) compared with control. We found a distinct increase in the mRNA expression of granzyme B and perforin, key markers of cytotoxic T cells. No differences in the marker genes of regulatory T cells were evident. Levels of the anti-inflammatory microRNAs miR-16 and miR-93 were significantly higher in the xenoperfused group than in the control group. CONCLUSIONS This study demonstrated that contact of human blood with pig endothelium activates cytotoxic T cells within the first few hours, indicating acute rejection processes. This is accompanied by upregulation of anti-inflammatory microRNAs, which may represent compensatory anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Roland Tomasi
- From the Department of Anesthesiology, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany.,From the Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rao JS, Matson AW, Taylor RT, Burlak C. Xenotransplantation Literature Update January/February 2021. Xenotransplantation 2021; 28:e12685. [PMID: 33884670 DOI: 10.1111/xen.12685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Joseph Sushil Rao
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, USA.,Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Anders W Matson
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, USA
| | - R Travis Taylor
- Department of Medical Microbiology and Immunology, University of Toledo Medical Center, Toledo, OH, USA
| | - Christopher Burlak
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
26
|
Thompson CP, Jagdale A, Walcott G, Iwase H, Foote JB, Cron RQ, Hara H, Cleveland DC, Cooper DKC. A perspective on the potential detrimental role of inflammation in pig orthotopic heart xenotransplantation. Xenotransplantation 2021; 28:e12687. [PMID: 33786912 DOI: 10.1111/xen.12687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/26/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
There is a critical shortage of deceased human donor organs for transplantation. The need is perhaps most acute in neonates and infants with life-threatening congenital heart disease, in whom mechanical support devices are largely unsuccessful. If orthotopic (life-supporting) heart transplantation (OHTx) were consistently successful in the genetically engineered pig-to-nonhuman primate (NHP) model, a clinical trial of bridging with a pig heart in such patients might be justified. However, the results of pig OHTx in NHPs have been mixed and largely poor. We hypothesise that a factor is the detrimental effects of the inflammatory response that is known to develop (a) during any surgical procedure that requires cardiopulmonary bypass, and (b) immediately after an NHP recipient is exposed to a pig xenograft. We suggest that the combination of these two inflammatory responses has a direct detrimental effect on pig heart graft function, but also, and possibly of more importance, on recipient baboon pulmonary function, which further impacts survival of the pig heart graft. In addition, the inflammatory response almost certainly adversely impacts the immune response to the graft. If our hypothesis is correct, the potential steps that could be taken to reduce the inflammatory response or its effects (with varying degrees of efficacy) include (a) white blood cell filtration, (b) complement depletion or inactivation, (c) immunosuppressive therapy, (d) high-dose corticosteroid therapy, (e) cytokine/chemokine-targeted therapy, (f) ultrafiltration or CytoSorb hemoperfusion, (g) reduction in the levels of endogenous catecholamines, (h) triiodothyronine therapy and (i) genetic engineering of the organ-source pig. Prevention of the inflammatory response, or attenuation of its effects, by judicious anti-inflammatory therapy may contribute not only to early survival of the recipient of a genetically engineered pig OHTx, but also to improved long-term pig heart graft survival. This would open the possibility of initiating a clinical trial of genetically engineered pig OHTx as a bridge to allotransplantation.
Collapse
Affiliation(s)
- Charles P Thompson
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Abhijit Jagdale
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory Walcott
- Department of Medicine/Cardiovascular Diseases, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hayato Iwase
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Randall Q Cron
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David C Cleveland
- Division of Cardiothoracic Surgery, Children's Hospital of Alabama, and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K C Cooper
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
27
|
Taghizadeh B, Ghavami L, Derakhshankhah H, Zangene E, Razmi M, Jaymand M, Zarrintaj P, Zarghami N, Jaafari MR, Moallem Shahri M, Moghaddasian A, Tayebi L, Izadi Z. Biomaterials in Valvular Heart Diseases. Front Bioeng Biotechnol 2020; 8:529244. [PMID: 33425862 PMCID: PMC7793990 DOI: 10.3389/fbioe.2020.529244] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 11/16/2020] [Indexed: 01/07/2023] Open
Abstract
Valvular heart disease (VHD) occurs as the result of valvular malfunction, which can greatly reduce patient's quality of life and if left untreated may lead to death. Different treatment regiments are available for management of this defect, which can be helpful in reducing the symptoms. The global commitment to reduce VHD-related mortality rates has enhanced the need for new therapeutic approaches. During the past decade, development of innovative pharmacological and surgical approaches have dramatically improved the quality of life for VHD patients, yet the search for low cost, more effective, and less invasive approaches is ongoing. The gold standard approach for VHD management is to replace or repair the injured valvular tissue with natural or synthetic biomaterials. Application of these biomaterials for cardiac valve regeneration and repair holds a great promise for treatment of this type of heart disease. The focus of the present review is the current use of different types of biomaterials in treatment of valvular heart diseases.
Collapse
Affiliation(s)
- Bita Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laleh Ghavami
- Laboratory of Biophysics and Molecular Biology, Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Zangene
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mahdieh Razmi
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Payam Zarrintaj
- Polymer Engineering Department, Faculty of Engineering, Urmia University, Urmia, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Matin Moallem Shahri
- Cardiology Department, Taleghani Trauma Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, United States
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Academic Center for Education, Culture and Research (ACECR), Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| |
Collapse
|
28
|
Reichart B, Längin M. On the way (my way) to clinical xenogeneic heart transplantation. Presented at the 15th biannual IXA meeting, Munich, October 11, 2019. Xenotransplantation 2020; 27:e12637. [DOI: 10.1111/xen.12637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023]
Affiliation(s)
- Bruno Reichart
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
| | - Matthias Längin
- Department of Anaesthesiology University Hospital LMU Munich Munich Germany
| |
Collapse
|
29
|
Längin M, Reichart B, Steen S, Sjöberg T, Paskevicius A, Liao Q, Qin G, Mokelke M, Mayr T, Radan J, Issl L, Buttgereit I, Ying J, Fresch AK, Panelli A, Egerer S, Bähr A, Kessler B, Milusev A, Sfriso R, Rieben R, Ayares D, Murray PJ, Ellgass R, Walz C, Klymiuk N, Wolf E, Abicht JM, Brenner P. Cold non-ischemic heart preservation with continuous perfusion prevents early graft failure in orthotopic pig-to-baboon xenotransplantation. Xenotransplantation 2020; 28:e12636. [PMID: 32841431 DOI: 10.1111/xen.12636] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/22/2020] [Accepted: 07/30/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Successful preclinical transplantations of porcine hearts into baboon recipients are required before commencing clinical trials. Despite years of research, over half of the orthotopic cardiac xenografts were lost during the first 48 hours after transplantation, primarily caused by perioperative cardiac xenograft dysfunction (PCXD). To decrease the rate of PCXD, we adopted a preservation technique of cold non-ischemic perfusion for our ongoing pig-to-baboon cardiac xenotransplantation project. METHODS Fourteen orthotopic cardiac xenotransplantation experiments were carried out with genetically modified juvenile pigs (GGTA1- KO/hCD46/hTBM) as donors and captive-bred baboons as recipients. Organ preservation was compared according to the two techniques applied: cold static ischemic cardioplegia (IC; n = 5) and cold non-ischemic continuous perfusion (CP; n = 9) with an oxygenated albumin-containing hyperoncotic cardioplegic solution containing nutrients, erythrocytes and hormones. Prior to surgery, we measured serum levels of preformed anti-non-Gal-antibodies. During surgery, hemodynamic parameters were monitored with transpulmonary thermodilution. Central venous blood gas analyses were taken at regular intervals to estimate oxygen extraction, as well as lactate production. After surgery, we measured troponine T and serum parameters of the recipient's kidney, liver and coagulation functions. RESULTS In porcine grafts preserved with IC, we found significantly depressed systolic cardiac function after transplantation which did not recover despite increasing inotropic support. Postoperative oxygen extraction and lactate production were significantly increased. Troponin T, creatinine, aspartate aminotransferase levels were pathologically high, whereas prothrombin ratios were abnormally low. In three of five IC experiments, PCXD developed within 24 hours. By contrast, all nine hearts preserved with CP retained fully preserved systolic function, none showed any signs of PCXD. Oxygen extraction was within normal ranges; serum lactate as well as parameters of organ functions were only mildly elevated. Preformed anti-non-Gal-antibodies were similar in recipients receiving grafts from either IC or CP preservation. CONCLUSIONS While standard ischemic cardioplegia solutions have been used with great success in human allotransplantation over many years, our data indicate that they are insufficient for preservation of porcine hearts transplanted into baboons: Ischemic storage caused severe impairment of cardiac function and decreased tissue oxygen supply, leading to multi-organ failure in more than half of the xenotransplantation experiments. In contrast, cold non-ischemic heart preservation with continuous perfusion reliably prevented early graft failure. Consistent survival in the perioperative phase is a prerequisite for preclinical long-term results after cardiac xenotransplantation.
Collapse
Affiliation(s)
- Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Stig Steen
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Trygve Sjöberg
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Audrius Paskevicius
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Qiuming Liao
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Guangqi Qin
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Maren Mokelke
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Tanja Mayr
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Julia Radan
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Lara Issl
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Ines Buttgereit
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Jiawei Ying
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Ann Kathrin Fresch
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Alessandro Panelli
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Stefanie Egerer
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Andrea Bähr
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Barbara Kessler
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Anastasia Milusev
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Riccardo Sfriso
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Reinhard Ellgass
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Walz
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Jan-Michael Abicht
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Paolo Brenner
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
30
|
Heterotopic Porcine Cardiac Xenotransplantation in the Intra-Abdominal Position in a Non-Human Primate Model. Sci Rep 2020; 10:10709. [PMID: 32612124 PMCID: PMC7329828 DOI: 10.1038/s41598-020-66430-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/19/2020] [Indexed: 12/27/2022] Open
Abstract
Heterotopic cardiac transplantation in the intra-abdominal position in a large animal model has been essential in the progression of the field of cardiac transplantation. Our group has over 10 years of experience in cardiac xenotransplantation with pig to baboon models, the longest xenograft of which survived over 900 days, with rejection only after reducing immunosuppression. This article aims to clarify our approach to this model in order to allow others to share success in long-term survival. Here, we demonstrate the approach to implantation of a cardiac graft into the intra-abdominal position in a baboon recipient for the study of transplantation and briefly highlight our model's ability to provide insight into not only xenotransplantation but across disciplines. We include details that have provided us with consistent success in this model; performance of the anastomoses, de-airing of the graft, implantation of a long-term telemetry device for invasive graft monitoring, and ideal geometric positioning of the heart and telemetry device in the limited space of the recipient abdomen. We additionally detail surveillance techniques to assess long-term graft function.
Collapse
|
31
|
Shu S, Ren J, Song J. Cardiac xenotransplantation: a promising way to treat advanced heart failure. Heart Fail Rev 2020; 27:71-91. [DOI: 10.1007/s10741-020-09989-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
32
|
A major advance toward clinical cardiac xenotransplantation. J Thorac Cardiovasc Surg 2020; 159:166-169. [PMID: 31204127 DOI: 10.1016/j.jtcvs.2019.04.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 01/14/2023]
|
33
|
Längin M, Konrad M, Reichart B, Mayr T, Vandewiele S, Postrach J, Mokelke M, Radan J, Brenner P, Bauer A, Abicht J. Hemodynamic evaluation of anesthetized baboons and piglets by transpulmonary thermodilution: Normal values and interspecies differences with respect to xenotransplantation. Xenotransplantation 2019; 27:e12576. [DOI: 10.1111/xen.12576] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/04/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Matthias Längin
- Department of Anaesthesiology University Hospital LMU Munich Munich Germany
| | | | - Bruno Reichart
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
| | - Tanja Mayr
- Department of Anaesthesiology University Hospital LMU Munich Munich Germany
| | - Stephanie Vandewiele
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
- Dr. von Haunersches Kinderspital Kinderklinik und Kinderpoliklinik der LMU München Munich Germany
| | - Johannes Postrach
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
- Privatklinik Dr. Robert Schindlbeck GmbH &Co KG Department of Medicine Herrsching am Ammersee Germany
| | - Maren Mokelke
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
| | - Julia Radan
- Transregional Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine LMU Munich Munich Germany
| | - Paolo Brenner
- Department of Cardiac Surgery University Hospital LMU Munich Munich Germany
| | - Andreas Bauer
- Department of Anaesthesiology and Intensive Care RoMed Klinikum Rosenheim Rosenheim Germany
| | - Jan‐Michael Abicht
- Department of Anaesthesiology University Hospital LMU Munich Munich Germany
| |
Collapse
|
34
|
Merkler AE, Chen ML, Parikh NS, Murthy SB, Yaghi S, Goyal P, Okin PM, Karas MG, Navi BB, Iadecola C, Kamel H. Association Between Heart Transplantation and Subsequent Risk of Stroke Among Patients With Heart Failure. Stroke 2019; 50:583-587. [PMID: 30744541 DOI: 10.1161/strokeaha.118.023622] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- It is uncertain whether heart transplantation decreases the risk of stroke. The objective of our study was to determine whether heart transplantation is associated with a decreased risk of subsequent stroke among patients with heart failure awaiting transplantation. Methods- We performed a retrospective cohort study using administrative data from New York, California, and Florida between 2005 and 2015. Individuals with heart failure awaiting heart transplantation were identified using previously validated International Classification of Diseases, Ninth Revision, Clinical Modification diagnosis codes for heart failure in combination with code V49.83 for awaiting organ transplant status. Individuals with prior stroke were excluded. Our primary exposure variable was heart transplantation, modeled as a time-varying covariate and defined by procedure code 37.51. The primary outcome was stroke, defined as the composite of ischemic and hemorrhagic stroke. Survival statistics were used to calculate stroke incidence, and Cox proportional hazards analysis was used to determine the association between heart transplantation and stroke while adjusting for demographics, stroke risk factors, Elixhauser comorbidities, and implantation of a left ventricular assist device. Results- We identified 7848 patients with heart failure awaiting heart transplantation, of whom 1068 (13.6%) underwent heart transplantation. During a mean follow-up of 2.7 years, we identified 428 strokes. The annual incidence of stroke was 0.7% (95% CI, 0.5%-1.0%) after heart transplantation versus 2.4% (95% CI, 2.2%-2.6%) among those awaiting heart transplantation. After adjustment for potential confounders, heart transplantation was associated with a lower risk of stroke (hazard ratio, 0.4; 95% CI, 0.2-0.6). Conclusions- Heart transplantation is associated with a decreased risk of stroke among patients with heart failure awaiting transplantation.
Collapse
Affiliation(s)
- Alexander E Merkler
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.).,Feil Family Brain and Mind Research Institute, Department of Neurology (A.E.M., S.B.M., B.B.N., C.I., H.K.)
| | - Monica L Chen
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.)
| | - Neal S Parikh
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.).,Department of Neurology, Columbia College of Physicians and Surgeons, New York, NY (N.S.P.)
| | - Santosh B Murthy
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.).,Feil Family Brain and Mind Research Institute, Department of Neurology (A.E.M., S.B.M., B.B.N., C.I., H.K.)
| | - Shadi Yaghi
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI (S.Y.)
| | - Parag Goyal
- Department of Cardiology, Weill Cornell Medical College, New York, NY (P.G., P.M.O., M.G.K.)
| | - Peter M Okin
- Department of Cardiology, Weill Cornell Medical College, New York, NY (P.G., P.M.O., M.G.K.)
| | - Maria G Karas
- Department of Cardiology, Weill Cornell Medical College, New York, NY (P.G., P.M.O., M.G.K.)
| | - Babak B Navi
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.).,Feil Family Brain and Mind Research Institute, Department of Neurology (A.E.M., S.B.M., B.B.N., C.I., H.K.)
| | - Costantino Iadecola
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.).,Feil Family Brain and Mind Research Institute, Department of Neurology (A.E.M., S.B.M., B.B.N., C.I., H.K.)
| | - Hooman Kamel
- From the Clinical and Translational Neuroscience Unit (A.E.M., M.L.C., N.S.P., S.B.M., B.B.N., C.I., H.K.).,Feil Family Brain and Mind Research Institute, Department of Neurology (A.E.M., S.B.M., B.B.N., C.I., H.K.)
| |
Collapse
|
35
|
DiChiacchio L, Singh AK, Lewis B, Zhang T, Hardy N, Pasrija C, Morales D, Odonkor P, Strauss E, Williams B, Deatrick KB, Kaczorowski DJ, Ayares D, Griffith BP, Bartlett ST, Mohiuddin MM. Early Experience With Preclinical Perioperative Cardiac Xenograft Dysfunction in a Single Program. Ann Thorac Surg 2019; 109:1357-1361. [PMID: 31589847 DOI: 10.1016/j.athoracsur.2019.08.090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/16/2019] [Accepted: 08/28/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Perioperative cardiac xenograft dysfunction (PCXD) was described by McGregor and colleagues as a major barrier to the translation of heterotopic cardiac xenotransplantation into the orthotopic position. It is characterized by graft dysfunction in the absence of rejection within 24 to 48 hours of transplantation. We describe our experience with PCXD at a single program. METHODS Orthotopic transplantation of genetically engineered pig hearts was performed in 6 healthy baboons. The immunosuppression regimen included induction by anti-CD20 monoclonal antibodies (mAb), thymoglobulin, cobra venom factor, and anti-CD40 mAb, and maintenance with anti-CD40 mAb, mycophenolate mofetil, and tapering doses of steroids. Telemetry was used to assess graft function. Extracorporeal membrane oxygenation was used to support 1 recipient. A full human clinical transplantation team was involved in these experiments and the procedure was performed by skilled transplantation surgeons. RESULTS A maximal survival of 40 hours was achieved in these experiments. The surgical procedures were uneventful, and all hearts were weaned from cardiopulmonary bypass without issue. Support with inotropes and vasopressors was generally required after separation from cardiopulmonary bypass. The cardiac xenografts performed well immediately, but within the first several hours they required increasing support and ultimately resulted in arrest despite maximal interventions. All hearts were explanted immediately; histology showed no signs of rejection. CONCLUSIONS Despite excellent surgical technique, uneventful weaning from cardiopulmonary bypass, and adequate initial function, orthotopic cardiac xenografts slowly fail within 24 to 48 hours without evidence of rejection. Modification of preservation techniques and minimizing donor organ ischemic time may be able to ameliorate PCXD.
Collapse
Affiliation(s)
- Laura DiChiacchio
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - Avneesh K Singh
- Department of Surgery, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland
| | - Billeta Lewis
- Department of Surgery, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland
| | - Tianshu Zhang
- Department of Surgery, University of Maryland Baltimore, School of Medicine, Baltimore, Maryland
| | - Naomi Hardy
- Department of Pathology, University of Maryland Medical Center, Baltimore, Maryland
| | - Chetan Pasrija
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - David Morales
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - Patrick Odonkor
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - Erik Strauss
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - Brittney Williams
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | | | - David J Kaczorowski
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | | | - Bartley P Griffith
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - Stephen T Bartlett
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland
| | - Muhammad M Mohiuddin
- Department of Surgery, University of Maryland Medical Center, Baltimore, Maryland.
| |
Collapse
|
36
|
DiChiacchio L, Singh AK, Chan JL, Shockcor NM, Zhang T, Lewis BG, Ayares D, Corcoran P, Horvath KA, Mohiuddin MM. Intra-Abdominal Heterotopic Cardiac Xenotransplantation: Pearls and Pitfalls. Front Cardiovasc Med 2019; 6:95. [PMID: 31404245 PMCID: PMC6669937 DOI: 10.3389/fcvm.2019.00095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/25/2019] [Indexed: 11/22/2022] Open
Abstract
Heterotopic cardiac xenotransplantation in the intra-abdominal position has been studied extensively in a pig-to-baboon model to define the optimal donor genetics and immunosuppressive regimen to prevent xenograft rejection. Extensive investigation using this model is a necessary stepping stone toward the development of a life-supporting animal model, with the ultimate goal of demonstrating suitability for clinical cardiac xenotransplantation trials. Aspects of surgical technique, pre- and post-operative care, graft monitoring, and minimization of infectious risk have all required refinement and optimization of heterotopic cardiac xenotransplantation over time. This review details non-immunologic obstacles relevant to this model described by our group and in the literature, as well as strategies that have been developed to address these specific challenges.
Collapse
Affiliation(s)
- Laura DiChiacchio
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Avneesh K. Singh
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Joshua L. Chan
- National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, United States
| | - Nicole M. Shockcor
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Tianshu Zhang
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | - Billeta G. Lewis
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| | | | - Philip Corcoran
- National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, United States
| | - Keith A. Horvath
- National Heart, Lung, Blood Institute, National Institute of Health, Bethesda, MD, United States
| | - Muhammad M. Mohiuddin
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, United States
| |
Collapse
|
37
|
Li KYC. Bioprosthetic Heart Valves: Upgrading a 50-Year Old Technology. Front Cardiovasc Med 2019; 6:47. [PMID: 31032263 PMCID: PMC6470412 DOI: 10.3389/fcvm.2019.00047] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/26/2019] [Indexed: 01/09/2023] Open
Abstract
Prosthetic heart valves have been commonly used to address the increasing prevalence of valvular heart disease. The ideal prosthetic heart valve substitute should closely mimic the characteristics of a normal native heart valve. Despite the development of various interventions, an exemplary valve replacement does not exist. This review provides an overview of the novel engineering valve designs and explores emergent immunologic insights into age-dependent structural valve degeneration (SVD).
Collapse
Affiliation(s)
- Kan Yan Chloe Li
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
38
|
Längin M, Mayr T, Reichart B, Michel S, Buchholz S, Guethoff S, Dashkevich A, Baehr A, Egerer S, Bauer A, Mihalj M, Panelli A, Issl L, Ying J, Fresch AK, Buttgereit I, Mokelke M, Radan J, Werner F, Lutzmann I, Steen S, Sjöberg T, Paskevicius A, Qiuming L, Sfriso R, Rieben R, Dahlhoff M, Kessler B, Kemter E, Kurome M, Zakhartchenko V, Klett K, Hinkel R, Kupatt C, Falkenau A, Reu S, Ellgass R, Herzog R, Binder U, Wich G, Skerra A, Ayares D, Kind A, Schönmann U, Kaup FJ, Hagl C, Wolf E, Klymiuk N, Brenner P, Abicht JM. Consistent success in life-supporting porcine cardiac xenotransplantation. Nature 2018; 564:430-433. [PMID: 30518863 DOI: 10.1038/s41586-018-0765-z] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 11/02/2018] [Indexed: 01/01/2023]
Abstract
Heart transplantation is the only cure for patients with terminal cardiac failure, but the supply of allogeneic donor organs falls far short of the clinical need1-3. Xenotransplantation of genetically modified pig hearts has been discussed as a potential alternative4. Genetically multi-modified pig hearts that lack galactose-α1,3-galactose epitopes (α1,3-galactosyltransferase knockout) and express a human membrane cofactor protein (CD46) and human thrombomodulin have survived for up to 945 days after heterotopic abdominal transplantation in baboons5. This model demonstrated long-term acceptance of discordant xenografts with safe immunosuppression but did not predict their life-supporting function. Despite 25 years of extensive research, the maximum survival of a baboon after heart replacement with a porcine xenograft was only 57 days and this was achieved, to our knowledge, only once6. Here we show that α1,3-galactosyltransferase-knockout pig hearts that express human CD46 and thrombomodulin require non-ischaemic preservation with continuous perfusion and control of post-transplantation growth to ensure long-term orthotopic function of the xenograft in baboons, the most stringent preclinical xenotransplantation model. Consistent life-supporting function of xenografted hearts for up to 195 days is a milestone on the way to clinical cardiac xenotransplantation7.
Collapse
Affiliation(s)
- Matthias Längin
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany.,Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Tanja Mayr
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany.,Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany.
| | - Sebastian Michel
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Buchholz
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Sonja Guethoff
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany.,Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Alexey Dashkevich
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Andrea Baehr
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Stefanie Egerer
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Andreas Bauer
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Maks Mihalj
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Alessandro Panelli
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Lara Issl
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Jiawei Ying
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Ann Kathrin Fresch
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Ines Buttgereit
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Maren Mokelke
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Julia Radan
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Fabian Werner
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Isabelle Lutzmann
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| | - Stig Steen
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Trygve Sjöberg
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Audrius Paskevicius
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Liao Qiuming
- Department of Cardiothoracic Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Riccardo Sfriso
- Department for BioMedical Research (DMBR), University of Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research (DMBR), University of Bern, Bern, Switzerland
| | - Maik Dahlhoff
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Barbara Kessler
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Elisabeth Kemter
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Mayuko Kurome
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Valeri Zakhartchenko
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Katharina Klett
- I. Medizinische Klinik, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Rabea Hinkel
- I. Medizinische Klinik, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Kupatt
- I. Medizinische Klinik, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Almuth Falkenau
- Institute of Veterinary Pathology, LMU Munich, Munich, Germany
| | - Simone Reu
- Institute of Pathology, Medical Faculty, LMU Munich, Munich, Germany
| | - Reinhard Ellgass
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Rudolf Herzog
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | | | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | | | | | - Christian Hagl
- Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Paolo Brenner
- Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany.,Department of Cardiac Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Jan-Michael Abicht
- Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany.,Transregional Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
39
|
Burdorf L, Harris D, Dahi S, Laird C, Zhang T, Ali F, Shah A, Thompson M, Braileanu G, Cheng X, Sievert E, Schwartz E, Sendil S, Parsell DM, Redding E, Phelps CJ, Ayares DL, Azimzadeh AM, Pierson RN. Thromboxane and histamine mediate PVR elevation during xenogeneic pig lung perfusion with human blood. Xenotransplantation 2018; 26:e12458. [PMID: 30175863 DOI: 10.1111/xen.12458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/29/2018] [Accepted: 07/20/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Elevated pulmonary vascular resistance (PVR), platelet adhesion, coagulation activation, and inflammation are prominent features of xenolung rejection. Here, we evaluate the role of thromboxane and histamine on PVR, and their contribution to other lung xenograft injury mechanisms. METHODS GalTKO.hCD46 single pig lungs were perfused ex vivo with fresh heparinized human blood: lungs were either treated with 1-Benzylimidazole (1-BIA) combined with histamine receptor blocker famotidine (n = 4) or diphenhydramine (n = 6), 1-BIA alone (n = 6) or were left untreated (n = 9). RESULTS Six of the nine control experiments (GalTKO.hCD46 untreated), "survived" until elective termination at 4 hours. Without treatment, initial PVR elevation within the first 30 minutes resolved partially over the following hour, and increased progressively during the final 2 hours of perfusion. In contrast, 1-BIA, alone or in addition to either antihistamine treatment, was associated with low stable PVR. Combined treatments significantly lowered the airway pressure when compared to untreated reference. Although platelet and neutrophil sequestration and coagulation cascade activation were not consistently altered by any intervention, increased terminal wet/dry weight ratio in untreated lungs was significantly blunted by combined treatments. CONCLUSION Combined thromboxane and histamine pathway blockade prevents PVR elevation and significantly inhibits loss of vascular barrier function when GalTKO.hCD46 lungs are perfused with human blood. Platelet activation and platelet and neutrophil sequestration persist in all groups despite efficient complement regulation, and appear to occur independent of thromboxane and histamine antagonism. Our work identifies thromboxane and histamine as key mediators of xenolung injury and defines those pathways as therapeutic targets to achieve successful xenolung transplantation.
Collapse
Affiliation(s)
- Lars Burdorf
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland.,Center for Transplantation Sciences and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Donald Harris
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Siamak Dahi
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Christopher Laird
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Tianshu Zhang
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Franchesca Ali
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Aakash Shah
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Mercedes Thompson
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Gheorghe Braileanu
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Xiangfei Cheng
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Evelyn Sievert
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Evan Schwartz
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Selin Sendil
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Dawn M Parsell
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Emily Redding
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland
| | - Carol J Phelps
- Center for Transplantation Sciences and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Agnes M Azimzadeh
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland.,Center for Transplantation Sciences and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard N Pierson
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, and VA Maryland Health Care System, Baltimore, Maryland.,Center for Transplantation Sciences and Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
40
|
Längin M, Panelli A, Reichart B, Kind A, Brenner P, Mayr T, Abicht JM. Perioperative Telemetric Monitoring in Pig-to-Baboon Heterotopic Thoracic Cardiac Xenotransplantation. Ann Transplant 2018; 23:491-499. [PMID: 30026460 PMCID: PMC6248073 DOI: 10.12659/aot.909522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Perioperative monitoring and hemodynamic management after heterotopic thoracic cardiac xenotransplantation is challenging due to 2 independently beating hearts. Telemetry allows continuous monitoring of hemodynamic parameters of both the donor and recipient hearts. We describe our experience and report on the validity of a telemetric system during and after surgery. Material/Methods Wireless telemetry transmitters were implanted in 3 baboons receiving porcine donor hearts. Left ventricular pressure and ECG were assessed from the donor heart, whereas aortic pressure and temperature were assessed from the recipient. Telemetric data were validated with invasive blood pressure measurements. Results Telemetric blood pressure was lower than invasive blood pressure. Intraoperatively, the probe in the graft’s left ventricle measured negative end-diastolic pressures. Telemetry allowed simple discrimination between donor’s and recipient’s heart rates. Body temperature was underestimated by telemetry. Telemetric monitoring facilitates recognition of graft arrhythmias and volume demand. Conclusions In heterotopic thoracic cardiac xenotransplantation, telemetric implants are useful tools to continuously monitor the animals’ hemodynamic parameters and to discriminate donor and recipient organs. Accuracy is sufficient for systemic pressure measurement, but perioperative use of left ventricular end-diastolic pressure as a surrogate parameter for graft function is not advisable. Temperature measurements by telemetry do not reflect body core temperature.
Collapse
Affiliation(s)
- Matthias Längin
- Department of Anaesthesiology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,Transregio Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Alessandro Panelli
- Transregio Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University (LMU), Munich, Germany.,Department of Cardiac Surgery, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| | - Bruno Reichart
- Transregio Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Paolo Brenner
- Transregio Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University (LMU), Munich, Germany.,Department of Cardiac Surgery, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| | - Tanja Mayr
- Department of Anaesthesiology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,Transregio Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Jan-Michael Abicht
- Department of Anaesthesiology, University Hospital, Ludwig Maximilian University (LMU), Munich, Germany.,Transregio Collaborative Research Center 127, Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
41
|
Wang L, Cooper DKC, Burdorf L, Wang Y, Iwase H. Overcoming Coagulation Dysregulation in Pig Solid Organ Transplantation in Nonhuman Primates: Recent Progress. Transplantation 2018; 102:1050-1058. [PMID: 29538262 PMCID: PMC7228622 DOI: 10.1097/tp.0000000000002171] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 01/17/2023]
Abstract
There has recently been considerable progress in the results of pig organ transplantation in nonhuman primates, largely associated with the availability of (i) pigs genetically engineered to overcome coagulation dysregulation, and (ii) novel immunosuppressive agents. The barriers of thrombotic microangiopathy and/or consumptive coagulation were believed to be associated with (i) activation of the graft vascular endothelial cells by a low level of antipig antibody binding and/or complement deposition and/or innate immune cell activity, and (ii) molecular incompatibilities between the nonhuman primate and pig coagulation-anticoagulation systems. The introduction of a human coagulation-regulatory transgene, for example, thrombomodulin, endothelial protein C receptor, into the pig vascular endothelial cells has contributed to preventing a procoagulant state from developing, resulting in a considerable increase in graft survival. In the heterotopic (non-life-supporting) heart transplant model, graft survival has increased from a maximum of 179 days in 2005 to 945 days. After life-supporting kidney transplantation, survival has been extended from 90 days in 2004 to 499 days. In view of the more complex coagulation dysfunction seen after pig liver and, particularly, lung transplantation, progress has been less dramatic, but the maximum survival of a pig liver has been increased from 7 days in 2010 to 29 days, and of a pig lung from 4 days in 2007 to 9 days. There is a realistic prospect that the transplantation of a kidney or heart, in combination with a conventional immunosuppressive regimen, will enable long-term recipient survival.
Collapse
Affiliation(s)
- Liaoran Wang
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham AL
- Second Affiliated Hospital, University of South China, Hengyang City, Hunan, China
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham AL
| | - Lars Burdorf
- Division of Cardiac Surgery, Department of Surgery, University of Maryland, Baltimore VAMC, Baltimore, MD
| | - Yi Wang
- Second Affiliated Hospital, University of South China, Hengyang City, Hunan, China
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham AL
| |
Collapse
|
42
|
Chan JL, Miller JG, Singh AK, Horvath KA, Corcoran PC, Mohiuddin MM. Consideration of appropriate clinical applications for cardiac xenotransplantation. Clin Transplant 2018; 32:e13330. [DOI: 10.1111/ctr.13330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Joshua L. Chan
- Cardiothoracic Surgery Research Program/National Heart; Lung and Blood Institute; National Institutes of Health; Bethesda MD USA
| | - Justin G. Miller
- Cardiothoracic Surgery Research Program/National Heart; Lung and Blood Institute; National Institutes of Health; Bethesda MD USA
| | - Avneesh K. Singh
- Cardiothoracic Surgery Research Program/National Heart; Lung and Blood Institute; National Institutes of Health; Bethesda MD USA
| | - Keith A. Horvath
- Cardiothoracic Surgery Research Program/National Heart; Lung and Blood Institute; National Institutes of Health; Bethesda MD USA
| | - Philip C. Corcoran
- Cardiothoracic Surgery Research Program/National Heart; Lung and Blood Institute; National Institutes of Health; Bethesda MD USA
| | - Muhammad M. Mohiuddin
- Cardiothoracic Surgery Research Program/National Heart; Lung and Blood Institute; National Institutes of Health; Bethesda MD USA
| |
Collapse
|
43
|
Patra C, Boccaccini A, Engel F. Vascularisation for cardiac tissue engineering: the extracellular matrix. Thromb Haemost 2017; 113:532-47. [DOI: 10.1160/th14-05-0480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
SummaryCardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 μm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial- based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.
Collapse
|
44
|
Porcine to Human Heart Transplantation: Is Clinical Application Now Appropriate? J Immunol Res 2017; 2017:2534653. [PMID: 29238731 PMCID: PMC5697125 DOI: 10.1155/2017/2534653] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/08/2017] [Indexed: 11/24/2022] Open
Abstract
Cardiac xenotransplantation (CXTx) is a promising solution to the chronic shortage of donor hearts. Recent advancements in immune suppression have greatly improved the survival of heterotopic CXTx, now extended beyond 2 years, and life-supporting kidney XTx. Advances in donor genetic modification (B4GALNT2 and CMAH mutations) with proven Gal-deficient donors expressing human complement regulatory protein(s) have also accelerated, reducing donor pig organ antigenicity. These advances can now be combined and tested in life-supporting orthotopic preclinical studies in nonhuman primates and immunologically appropriate models confirming their efficacy and safety for a clinical CXTx program. Preclinical studies should also allow for organ rejection to develop xenospecific assays and therapies to reverse rejection. The complexity of future clinical CXTx presents a substantial and unique set of regulatory challenges which must be addressed to avoid delay; however, dependent on these prospective life-supporting preclinical studies in NHPs, it appears that the scientific path forward is well defined and the era of clinical CXTx is approaching.
Collapse
|
45
|
Mayr T, Bauer A, Reichart B, Guethoff S, Schoenmann U, Längin M, Panelli A, Kind A, Brenner P, Abicht JM. Hemodynamic and perioperative management in two different preclinical pig-to-baboon cardiac xenotransplantation models. Xenotransplantation 2017; 24. [DOI: 10.1111/xen.12295] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/29/2016] [Accepted: 01/26/2017] [Indexed: 12/01/2022]
Affiliation(s)
- Tanja Mayr
- Department of Anaesthesiology; Ludwig Maximilian University; Munich Germany
| | - Andreas Bauer
- Department of Anaesthesiology; Ludwig Maximilian University; Munich Germany
| | - Bruno Reichart
- Walter Brendel Centre of Experimental Medicine; Ludwig Maximilian University; Munich Germany
| | - Sonja Guethoff
- Walter Brendel Centre of Experimental Medicine; Ludwig Maximilian University; Munich Germany
| | | | - Matthias Längin
- Department of Anaesthesiology; Ludwig Maximilian University; Munich Germany
| | - Alessandro Panelli
- Walter Brendel Centre of Experimental Medicine; Ludwig Maximilian University; Munich Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology; School of Life Sciences Weihenstephan; Technical University of Munich; Munich Germany
| | - Paolo Brenner
- Department of Cardiac Surgery; Ludwig Maximilian University; Munich Germany
| | - Jan-Michael Abicht
- Department of Anaesthesiology; Ludwig Maximilian University; Munich Germany
| |
Collapse
|
46
|
Abicht JM, Kourtzelis I, Reichart B, Koutsogiannaki S, Primikyri A, Lambris JD, Chavakis T, Holdt L, Kind A, Guethoff S, Mayr T. Complement C3 inhibitor Cp40 attenuates xenoreactions in pig hearts perfused with human blood. Xenotransplantation 2017; 24:10.1111/xen.12262. [PMID: 27677785 PMCID: PMC5358808 DOI: 10.1111/xen.12262] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/11/2016] [Accepted: 08/12/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND The complement system plays a crucial role in acute xenogeneic reactions after cardiac transplantation. We used an ex vivo perfusion model to investigate the effect of Cp40, a compstatin analog and potent inhibitor of complement at the level of C3. METHODS Fifteen wild-type pig hearts were explanted, cardiopleged, and reperfused ex vivo after 150 minutes of cold ischemia. Hearts were challenged in a biventricular working heart mode to evaluate cardiac perfusion and function. In the treatment group (n=5), the complement cascade was blocked at the level of C3 using Cp40, using diluted human blood. Untreated human and porcine blood was used for controls. RESULTS Throughout the perfusion, C3 activation was inhibited when Cp40 was used (mean of all time points: 1.11 ± 0.34% vs 3.12 ± 0.48% control activation; P<.01). Compared to xenoperfused controls, the cardiac index improved significantly in the treated group (6.5 ± 4.2 vs 3.5 ± 4.8 mL/min/g; P=.03, 180 minutes perfusion), while the concentration of lactate dehydrogenase as a maker for cell degradation was reduced in the perfusate (583 ± 187 U/mL vs 2108 ± 1145 U/mL, P=.02). Histological examination revealed less hemorrhage and edema, and immunohistochemistry confirmed less complement fragment deposition than in untreated xenoperfused controls. CONCLUSIONS Cp40 efficiently prevents C3 activation of the complement system, resulting in reduced cell damage and preserved function in wild-type porcine hearts xenoperfused ex vivo. We suggest that this compstatin analog, which blocks all main pathways of complement activation, could be a beneficial perioperative treatment in preclinical and in future clinical xenotransplantation.
Collapse
Affiliation(s)
- Jan-Michael Abicht
- Department of Anaesthesiology, Ludwig Maximilian University, Munich, Germany
| | - Ioannis Kourtzelis
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Germany
| | - Bruno Reichart
- Walter-Brendel-Centre, Ludwig Maximilian University Munich, Germany
| | - Sophia Koutsogiannaki
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Alexandra Primikyri
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - John D. Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Medical Faculty, Technische Universität Dresden, Germany
| | - Lesca Holdt
- Institute of Laboratory Medicine of Ludwig Maximilian University, Munich, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Germany
| | - Sonja Guethoff
- Department of Cardiovascular Surgery, Ludwig Maximilian University, Munich, Germany
| | - Tanja Mayr
- Department of Anaesthesiology, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
47
|
Iwase H, Ekser B, Hara H, Ezzelarab MB, Long C, Thomson AW, Ayares D, Cooper DKC. Thyroid hormone: relevance to xenotransplantation. Xenotransplantation 2016; 23:293-9. [PMID: 27374212 DOI: 10.1111/xen.12243] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/23/2016] [Indexed: 12/01/2022]
Abstract
BACKGROUND It has been well documented that the level of serum/plasma free triiodothyronine (fT3) falls rapidly following brain death or during certain surgical procedures, for example, heart surgery carried out on cardiopulmonary bypass. The level in patients following cardiopulmonary bypass usually recovers within 2 days. METHODS We have measured serum fT3 in healthy naïve baboons (n = 31), healthy naïve monkeys (n = 5), and after pig-to-baboon heterotopic heart xenotransplantation (xenoTx) (Group 1, n = 9), orthotopic liver xenoTx (Group 2, n = 10), artery patch xenoTx (Group 3, n = 9), and in monkey-to-monkey heterotopic heart alloTx (Group 4, n = 5). RESULTS The mean level of fT3 in healthy naïve baboons was 3.1 ± 0.9 pg/ml and in healthy naïve monkeys was 2.6 ± 0.3 pg/ml. Following pig heart, liver, and artery patch xenoTx and monkey heart alloTx, there was an immediate rapid fall in fT3 level. Recovery of fT3 was more rapid in Groups 3 and 4 than in Groups 1 and 2. In Group 1, within 4 days fT3 had recovered, but only to the lower limit of normal range, where it remained throughout follow-up (for up to 42 days). In Group 2, no recovery was seen during the 7 days of follow-up. In immunosuppressed baboons with pig patch grafts that received IL-6R blockade (n = 2), the fT3 tended to rise higher than in those that received no IL-6R blockade (n = 6). CONCLUSIONS Following operative procedures, there is a dramatic fall in serum fT3 levels. The persistent low level of fT3 after pig heart and liver xenoTx may be associated with a continuing inflammatory state. We suggest that consideration should be given to the replacement of T3 therapy to maintain normal fT3 levels, particularly in nonhuman primates undergoing orthotopic pig heart or liver xenoTx.
Collapse
Affiliation(s)
- Hayato Iwase
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Burcin Ekser
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hidetaka Hara
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohamed B Ezzelarab
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cassandra Long
- Thomas E Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Angus W Thomson
- 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
| |
Collapse
|
48
|
Parmaksiz M, Dogan A, Odabas S, Elçin AE, Elçin YM. Clinical applications of decellularized extracellular matrices for tissue engineering and regenerative medicine. Biomed Mater 2016; 11:022003. [DOI: 10.1088/1748-6041/11/2/022003] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
TRAN HLB, DINH TTH, NGUYEN MTN, TO QM, PHAM ATT. Preparation and characterization of acellular porcinepericardium for cardiovascular surgery. Turk J Biol 2016. [DOI: 10.3906/biy-1510-44] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
50
|
Abicht J, Mayr T, Reichart B, Buchholz S, Werner F, Lutzmann I, Schmoeckel M, Bauer A, Thormann M, Langenmayer M, Herbach N, Pohla H, Herzog R, McGregor CGA, Ayares D, Wolf E, Klymiuk N, Baehr A, Kind A, Hagl C, Ganswindt U, Belka C, Guethoff S, Brenner P. Pre‐clinical heterotopic intrathoracic heart xenotransplantation: a possibly useful clinical technique. Xenotransplantation 2015; 22:427-42. [DOI: 10.1111/xen.12213] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/19/2015] [Indexed: 01/24/2023]
Affiliation(s)
- Jan‐Michael Abicht
- Department of Anaesthesiology Ludwig‐Maximilian University Munich Germany
- Transregio Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine Ludwig Maximilian University Munich Germany
| | - Tanja Mayr
- Department of Anaesthesiology Ludwig‐Maximilian University Munich Germany
- Transregio Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine Ludwig Maximilian University Munich Germany
| | - Bruno Reichart
- Transregio Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine Ludwig Maximilian University Munich Germany
| | - Stefan Buchholz
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| | - Fabian Werner
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| | - Isabelle Lutzmann
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| | - Michael Schmoeckel
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
- Department of Cardiac Surgery Asklepios Klinik St Georg Hamburg Germany
| | - Andreas Bauer
- Department of Anaesthesiology Ludwig‐Maximilian University Munich Germany
| | - Michael Thormann
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| | - Martin Langenmayer
- Institute of Veterinary Pathology Ludwig Maximilian University Munich Germany
| | - Nadja Herbach
- Institute of Veterinary Pathology Ludwig Maximilian University Munich Germany
| | - Heike Pohla
- Tumor Immunology Laboratory LIFE Center Ludwig Maximilian University Munich Germany
| | - Rudolf Herzog
- Transregio Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine Ludwig Maximilian University Munich Germany
| | | | | | - Eckhard Wolf
- Department of Molecular Animal Breeding and Biotechnology Ludwig Maximilian University Munich Germany
| | - Nikolai Klymiuk
- Department of Molecular Animal Breeding and Biotechnology Ludwig Maximilian University Munich Germany
| | - Andrea Baehr
- Department of Molecular Animal Breeding and Biotechnology Ludwig Maximilian University Munich Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology School of Life Sciences Weihenstephan Technical University of Munich Germany
| | - Christian Hagl
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| | - Ute Ganswindt
- Department of Radiation Oncology Ludwig Maximilian University Munich Germany
| | - Claus Belka
- Department of Radiation Oncology Ludwig Maximilian University Munich Germany
| | - Sonja Guethoff
- Transregio Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine Ludwig Maximilian University Munich Germany
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| | - Paolo Brenner
- Transregio Collaborative Research Center 127 Walter Brendel Centre of Experimental Medicine Ludwig Maximilian University Munich Germany
- Department of Cardiovascular Surgery Ludwig Maximilian University Munich Germany
| |
Collapse
|