1
|
Boiko JR, Hill GR. Chronic Graft-versus-host Disease: Immune Insights, Therapeutic Advances, and Parallels for Solid Organ Transplantation. Transplantation 2024:00007890-990000000-00959. [PMID: 39682018 DOI: 10.1097/tp.0000000000005298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Chronic graft-versus-host disease remains a frequent and morbid outcome of allogeneic hematopoietic cell transplantation, in which the donor-derived immune system attacks healthy recipient tissue. Preceding tissue damage mediated by chemoradiotherapy and alloreactive T cells compromise central and peripheral tolerance mechanisms, leading to aberrant donor T cell and germinal center B cell differentiation, culminating in pathogenic macrophage infiltration and differentiation in a target tissue, with ensuant fibrosis. This process results in a heterogeneous clinical syndrome with significant morbidity and mortality, frequently requiring prolonged therapy. In this review, we discuss the processes that interrupt immune tolerance, the subsequent clinical manifestations, and new Food and Drug Administration-approved therapeutic approaches that have been born from a greater understanding of disease pathogenesis in preclinical systems, linking to parallel processes following solid organ transplantation.
Collapse
Affiliation(s)
- Julie R Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
2
|
Santos E, Lucisano G, Dor FJMF, Willicombe M. Comparative outcomes of DSA positive, crossmatch negative living donor kidney transplants versus remaining on the waitlist for an HLA compatible deceased donor. Transpl Immunol 2024; 86:102098. [PMID: 39074764 DOI: 10.1016/j.trim.2024.102098] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION The clinical relevance of preformed donor specific antibodies in the setting of a negative crossmatch (DSA + XM-) remains controversial. In this study we investigate the outcomes of patients with a DSA + XM- living donor (LDi) who proceeded with an HLA-incompatible (HLAi) transplant compared with those who waited for an HLA-compatible deceased donor (DDc). MATERIALS AND METHODS We investigated 359 patients on the transplant waiting list who had at least one potential HLAi living donor, from which 203 DSA + XM- pairs were identified and outcomes analysed. RESULTS Out of 203 patients, 96 (47.3%) received a LD transplant: 52/96 (54.2%) a LDi, and 44/96 (45.8%) an alternative compatible LD. In addition, 107 patients out of 203(52.7%) waited for a DDc, of which 47(43.9%) were subsequently transplanted. Our adjusted analysis showed that the LDi transplantation did not offer a superior patient survival over waiting for a DDc transplant. For those transplanted, there was no difference in patient (p = 0.065) or death censored allograft survival (p = 0.37) between DDc and LDi recipients. However, there was a higher incidence of acute allograft rejection (p = 0.043) and antibody-mediated rejection (p = 0.005) in the LDi group. Having a high pre-transplant calculated reaction frequency and preformed DSA to both class I and class II antigens were associated with inferior outcomes in the LDi transplants. CONCLUSIONS Given the lack of difference in allograft survival between LDi and DDc transplants, in the absence of an alternative compatible living donor, proceeding with a LDi should be supported despite a higher rejection risk, providing individual risk assessment and shared decision making is undertaken.
Collapse
Affiliation(s)
- Eva Santos
- Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK.
| | - Gaetano Lucisano
- Histocompatibility and Immunogenetics, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Frank J M F Dor
- Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - Michelle Willicombe
- Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK; Histocompatibility and Immunogenetics, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK; Centre for Inflammatory Disease, Division of Immunology and Inflammation, Department of Medicine, Imperial College London, UK
| |
Collapse
|
3
|
Aburahma K, de Manna ND, Kuehn C, Salman J, Greer M, Ius F. Pushing the Survival Bar Higher: Two Decades of Innovation in Lung Transplantation. J Clin Med 2024; 13:5516. [PMID: 39337005 PMCID: PMC11432129 DOI: 10.3390/jcm13185516] [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: 08/23/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Survival after lung transplantation has significantly improved during the last two decades. The refinement of the already existing extracorporeal life support (ECLS) systems, such as extracorporeal membrane oxygenation (ECMO), and the introduction of new techniques for donor lung optimization, such as ex vivo lung perfusion (EVLP), have allowed the extension of transplant indication to patients with end-stage lung failure after acute respiratory distress syndrome (ARDS) and the expansion of the donor organ pool, due to the better evaluation and optimization of extended-criteria donor (ECD) lungs and of donors after circulatory death (DCD). The close monitoring of anti-HLA donor-specific antibodies (DSAs) has allowed the early recognition of pulmonary antibody-mediated rejection (AMR), which requires a completely different treatment and has a worse prognosis than acute cellular rejection (ACR). As such, the standardization of patient selection and post-transplant management has significantly contributed to this positive trend, especially at high-volume centers. This review focuses on lung transplantation after ARDS, on the role of EVLP in lung donor expansion, on ECMO as a principal cardiopulmonary support system in lung transplantation, and on the diagnosis and therapy of pulmonary AMR.
Collapse
Affiliation(s)
- Khalil Aburahma
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Nunzio Davide de Manna
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Kuehn
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
| | - Jawad Salman
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
| | - Mark Greer
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, 30625 Hannover, Germany
| | - Fabio Ius
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Lung Research (DZL/BREATH), 35392 Hannover, Germany
| |
Collapse
|
4
|
van Zyl M, Cramer E, Sanders JSF, Leuvenink HGD, Lisman T, van Rooy MJ, Hillebrands JL. The role of neutrophil extracellular trap formation in kidney transplantation: Implications from donors to the recipient. Am J Transplant 2024; 24:1547-1557. [PMID: 38719094 DOI: 10.1016/j.ajt.2024.04.018] [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: 01/19/2024] [Revised: 03/19/2024] [Accepted: 04/05/2024] [Indexed: 05/23/2024]
Abstract
Kidney transplantation remains the gold standard for patients with end-stage renal disease, but severe donor organ shortage has led to long waiting lists. The utilization of expanded criteria donor kidneys within the category of deceased donors has enlarged the pool of available kidneys for transplantation; however, these grafts often have an increased risk for delayed graft function or reduced graft survival following transplantation. During brain or circulatory death, neutrophils are recruited to the vascular beds of kidneys where a proinflammatory microenvironment might prime the formation of neutrophil extracellular traps (NETs), web-like structures, containing proteolytic enzymes, DNA, and histones. NETs are known to cause tissue damage and specifically endothelial damage while activating other systems such as coagulation and complement, contributing to tissue injury and an unfavorable prognosis in various diseases. In lung transplantation and kidney transplantation studies, NETs have also been associated with primary graft dysfunction or rejection. In this review, the role that NETs might play across the different phases of transplantation, already initiated in the donor, during preservation, and in the recipient, will be discussed. Based on current knowledge, NETs might be a promising therapeutic target to improve graft outcomes.
Collapse
Affiliation(s)
- Maryna van Zyl
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands; Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Elodie Cramer
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan-Stephan F Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, the Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Ton Lisman
- Department of Surgery, University Medical Center Groningen, Groningen, the Netherlands
| | - Mia-Jeanne van Rooy
- Department of Physiology, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
5
|
Berg T, Aehling NF, Bruns T, Welker MW, Weismüller T, Trebicka J, Tacke F, Strnad P, Sterneck M, Settmacher U, Seehofer D, Schott E, Schnitzbauer AA, Schmidt HH, Schlitt HJ, Pratschke J, Pascher A, Neumann U, Manekeller S, Lammert F, Klein I, Kirchner G, Guba M, Glanemann M, Engelmann C, Canbay AE, Braun F, Berg CP, Bechstein WO, Becker T, Trautwein C. S2k-Leitlinie Lebertransplantation der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) und der Deutschen Gesellschaft für Allgemein- und Viszeralchirurgie (DGAV). ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:1397-1573. [PMID: 39250961 DOI: 10.1055/a-2255-7246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Affiliation(s)
- Thomas Berg
- Bereich Hepatologie, Medizinischen Klinik II, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Niklas F Aehling
- Bereich Hepatologie, Medizinischen Klinik II, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Tony Bruns
- Medizinische Klinik III, Universitätsklinikum Aachen, Aachen, Deutschland
| | - Martin-Walter Welker
- Medizinische Klinik I Gastroent., Hepat., Pneum., Endokrin. Universitätsklinikum Frankfurt, Frankfurt, Deutschland
| | - Tobias Weismüller
- Klinik für Innere Medizin - Gastroenterologie und Hepatologie, Vivantes Humboldt-Klinikum, Berlin, Deutschland
| | - Jonel Trebicka
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Deutschland
| | - Frank Tacke
- Charité - Universitätsmedizin Berlin, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Campus Virchow-Klinikum (CVK) und Campus Charité Mitte (CCM), Berlin, Deutschland
| | - Pavel Strnad
- Medizinische Klinik III, Universitätsklinikum Aachen, Aachen, Deutschland
| | - Martina Sterneck
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Hamburg, Hamburg, Deutschland
| | - Utz Settmacher
- Klinik für Allgemein-, Viszeral- und Gefäßchirurgie, Universitätsklinikum Jena, Jena, Deutschland
| | - Daniel Seehofer
- Klinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Eckart Schott
- Klinik für Innere Medizin II - Gastroenterologie, Hepatologie und Diabetolgie, Helios Klinikum Emil von Behring, Berlin, Deutschland
| | | | - Hartmut H Schmidt
- Klinik für Gastroenterologie und Hepatologie, Universitätsklinikum Essen, Essen, Deutschland
| | - Hans J Schlitt
- Klinik und Poliklinik für Chirurgie, Universitätsklinikum Regensburg, Regensburg, Deutschland
| | - Johann Pratschke
- Chirurgische Klinik, Charité Campus Virchow-Klinikum - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Andreas Pascher
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Universitätsklinikum Münster, Münster, Deutschland
| | - Ulf Neumann
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Universitätsklinikum Essen, Essen, Deutschland
| | - Steffen Manekeller
- Klinik und Poliklinik für Allgemein-, Viszeral-, Thorax- und Gefäßchirurgie, Universitätsklinikum Bonn, Bonn, Deutschland
| | - Frank Lammert
- Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
| | - Ingo Klein
- Chirurgische Klinik I, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Gabriele Kirchner
- Klinik und Poliklinik für Chirurgie, Universitätsklinikum Regensburg und Innere Medizin I, Caritaskrankenhaus St. Josef Regensburg, Regensburg, Deutschland
| | - Markus Guba
- Klinik für Allgemeine, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Universitätsklinikum München, München, Deutschland
| | - Matthias Glanemann
- Klinik für Allgemeine, Viszeral-, Gefäß- und Kinderchirurgie, Universitätsklinikum des Saarlandes, Homburg, Deutschland
| | - Cornelius Engelmann
- Charité - Universitätsmedizin Berlin, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Campus Virchow-Klinikum (CVK) und Campus Charité Mitte (CCM), Berlin, Deutschland
| | - Ali E Canbay
- Medizinische Klinik, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Deutschland
| | - Felix Braun
- Klinik für Allgemeine Chirurgie, Viszeral-, Thorax-, Transplantations- und Kinderchirurgie, Universitätsklinikum Schlewswig-Holstein, Kiel, Deutschland
| | - Christoph P Berg
- Innere Medizin I Gastroenterologie, Hepatologie, Infektiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - Wolf O Bechstein
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Frankfurt, Frankfurt, Deutschland
| | - Thomas Becker
- Klinik für Allgemeine Chirurgie, Viszeral-, Thorax-, Transplantations- und Kinderchirurgie, Universitätsklinikum Schlewswig-Holstein, Kiel, Deutschland
| | | |
Collapse
|
6
|
Messika J, Belousova N, Parquin F, Roux A. Antibody-Mediated Rejection in Lung Transplantation: Diagnosis and Therapeutic Armamentarium in a 21st Century Perspective. Transpl Int 2024; 37:12973. [PMID: 39170865 PMCID: PMC11336419 DOI: 10.3389/ti.2024.12973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024]
Abstract
Humoral immunity is a major waypoint towards chronic allograft dysfunction in lung transplantation (LT) recipients. Though allo-immunization and antibody-mediated rejection (AMR) are well-known entities, some diagnostic gaps need to be addressed. Morphological analysis could be enhanced by digital pathology and artificial intelligence-based companion tools. Graft transcriptomics can help to identify graft failure phenotypes or endotypes. Donor-derived cell free DNA is being evaluated for graft-loss risk stratification and tailored surveillance. Preventative therapies should be tailored according to risk. The donor pool can be enlarged for candidates with HLA sensitization, with strategies combining plasma exchange, intravenous immunoglobulin and immune cell depletion, or with emerging or innovative therapies such as imlifidase or immunoadsorption. In cases of insufficient pre-transplant desensitization, the effects of antibodies on the allograft can be prevented by targeting the complement cascade, although evidence for this strategy in LT is limited. In LT recipients with a humoral response, strategies are combined, including depletion of immune cells (plasmapheresis or immunoadsorption), inhibition of immune pathways, or modulation of the inflammatory cascade, which can be achieved with photopheresis. Altogether, these innovative techniques offer promising perspectives for LT recipients and shape the 21st century's armamentarium against AMR.
Collapse
Affiliation(s)
- Jonathan Messika
- Thoracic Intensive Care Unit, Foch Hospital, Suresnes, France
- Physiopathology and Epidemiology of Respiratory Diseases, UMR1152 INSERM and Université de Paris, Paris, France
- Paris Transplant Group, Paris, France
| | - Natalia Belousova
- Paris Transplant Group, Paris, France
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
| | - François Parquin
- Thoracic Intensive Care Unit, Foch Hospital, Suresnes, France
- Paris Transplant Group, Paris, France
| | - Antoine Roux
- Paris Transplant Group, Paris, France
- Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department, Foch Hospital, Suresnes, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| |
Collapse
|
7
|
Xu Q, Zeevi A, Ganoza A, Cruz RJ, Mazariegos GV. Current approaches for risk assessment of intestinal transplant patients: A view from the histocompatibility laboratory. Hum Immunol 2024; 85:110768. [PMID: 38433035 DOI: 10.1016/j.humimm.2024.110768] [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: 01/25/2024] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
Despite its recent decline in volumes, intestinal transplantation remains an important option for patients with irreversible intestinal failures. The long-term outcome of an intestinal transplant has stagnated. The major cause of graft loss is rejection, resulting from mismatches in human leukocyte antigens (HLA) and the presence of antibodies to mismatched donor-specific HLA antigens (DSA). Literature has reported that DSAs, either preformed before transplantation or developed de novo after transplantation, are harmful to intestinal grafts, especially for those without combined liver grafts. A comprehensive assessment of DSA by the histocompatibility laboratory is critical for successful intestinal transplantation and its long-term survival. This paper briefly reviews the history and current status of different methods for detecting DSA and their clinical applications in intestinal transplantation. The focus is on applying different antibody assays to manage immunologically challenging intestinal transplant patients before and after transplantation. A clinical case is presented to illustrate the complexity of HLA tests and the necessity of multiple assays. The review of risk assessment by the histocompatibility laboratory also highlights the need for close interaction between the laboratory and the intestinal transplant program.
Collapse
Affiliation(s)
- Qingyong Xu
- Department of Pathology, University of Pittsburgh, USA.
| | - Adriana Zeevi
- Department of Pathology, University of Pittsburgh, USA
| | | | - Ruy J Cruz
- Department of Surgery, University of Pittsburgh, USA; Gastrointestinal Rehabilitation and Transplant Center, Starzl Transplantation Institute, USA
| | - George V Mazariegos
- Department of Surgery, University of Pittsburgh, USA; Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh, USA
| |
Collapse
|
8
|
Ma N, Wu WB, Zhao XY, Xu LP, Zhang XH, Wang Y, Mo XD, Zhang YY, Zhao XS, Sun YQ, Cheng YF, Liu KY, Chang YJ, Huang XJ. Targeting T FH cells is a novel approach for donor-specific antibody desensitization of allograft candidates: an in vitro and in vivo study. Haematologica 2024; 109:1233-1246. [PMID: 37822236 PMCID: PMC10985458 DOI: 10.3324/haematol.2023.283698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
The presence of donor-specific antibodies (DSA) are associated with graft failure either following human leukocyte antigen (HLA)-mismatched allogeneic stem cell transplantation or after organ transplantation. Although targeting B cells and plasma cells have been used for desensitization, there have been reports of failure. T-follicular helper (Tfh) cells assist B cells in differentiating into antibody-secreting plasma cells. We used haploidentical allograft as a platform to investigate the possibility of targeting Tfh cells to desensitize DSA. The quantities of circulating Tfh (cTfh) cell subsets in allograft candidates were abnormal, and these cells, including the cTfh2 and cTfhem cell subsets, were positively related to the production of anti-HLA antibodies. Ex vivo experiments showed that the cTfh cells of anti-HLA antibody-positive allograft candidates could induce B cells to differentiate into DSA-producing plasmablasts. The immune synapse could be involved in the assistance of cTfh cells to B cells in antibody production. In vitro experiments and in vivo clinical pilot studies indicated that targeting cTfh cells with sirolimus can inhibit their auxiliary function in assisting B cells. Ex vivo and in vivo studies demonstrated the effect of sirolimus and rituximab on DSA desensitization compared with either sirolimus or rituximab alone (60%, 43.75%, and 30%, respectively). Our findings provide new insight into the role of Tfh cells in the pathogenesis of DSA production in HLA-mismatched transplant candidates. Our data also indicate that targeting Tfh cells is a novel strategy for DSA desensitization and combination of sirolimus and rituximab might be a potential therapy. The prospective cohort of this study is registered at http://www.chictr.org.cn as #ChiCTR-OPC-15006672.
Collapse
Affiliation(s)
- Ning Ma
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Wei-Bing Wu
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Xiang-Yu Zhao
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Lan-Ping Xu
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Xiao-Hui Zhang
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Yu Wang
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Xiao-Dong Mo
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Yuan-Yuan Zhang
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Xiao-Su Zhao
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Yu-Qian Sun
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Yi-Fei Cheng
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Kai-Yan Liu
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044
| | - Ying-Jun Chang
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044.
| | - Xiao-Jun Huang
- Peking University People's Hospital and Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, China; Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, 2019RU029, Beijing.
| |
Collapse
|
9
|
Cleary SJ, Seo Y, Tian JJ, Kwaan N, Bulkley DP, Bentlage AE, Vidarsson G, Boilard É, Spirig R, Zimring JC, Looney MR. IgG hexamers initiate complement-dependent acute lung injury. J Clin Invest 2024; 134:e178351. [PMID: 38530369 PMCID: PMC11142733 DOI: 10.1172/jci178351] [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: 12/15/2023] [Accepted: 03/20/2024] [Indexed: 03/28/2024] Open
Abstract
Antibodies can initiate lung injury in a variety of disease states such as autoimmunity, in reactions to transfusions, or after organ transplantation, but the key factors determining in vivo pathogenicity of injury-inducing antibodies are unclear. Harmful antibodies often activate the complement cascade. A model for how IgG antibodies trigger complement activation involves interactions between IgG Fc domains driving the assembly of IgG hexamer structures that activate C1 complexes. The importance of IgG hexamers in initiating injury responses was not clear, so we tested their relevance in a mouse model of alloantibody- and complement-mediated acute lung injury. We used 3 approaches to block alloantibody hexamerization (antibody carbamylation, the K439E Fc mutation, or treatment with domain B from staphylococcal protein A), all of which reduced acute lung injury. Conversely, Fc mutations promoting spontaneous hexamerization made a harmful alloantibody into a more potent inducer of acute lung injury and rendered an innocuous alloantibody pathogenic. Treatment with a recombinant Fc hexamer "decoy" therapeutic protected mice from lung injury, including in a model with transgenic human FCGR2A expression that exacerbated pathology. These results indicate an in vivo role of IgG hexamerization in initiating acute lung injury and the potential for therapeutics that inhibit or mimic hexamerization to treat antibody-mediated diseases.
Collapse
Affiliation(s)
| | | | | | | | - David P. Bulkley
- Department of Biochemistry and Biophysics, UCSF, San Francisco, California, USA
| | | | | | - Éric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec – Université Laval, Québec, Quebec, Canada
| | - Rolf Spirig
- CSL Behring, Research, CSL Behring Biologics Research Center, Bern, Switzerland
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
10
|
Raynaud M, Al-Awadhi S, Louis K, Zhang H, Su X, Goutaudier V, Wang J, Demir Z, Wei Y, Truchot A, Bouquegneau A, Del Bello A, Bailly É, Lombardi Y, Maanaoui M, Giarraputo A, Naser S, Divard G, Aubert O, Murad MH, Wang C, Liu L, Bestard O, Naesens M, Friedewald JJ, Lefaucheur C, Riella L, Collins G, Ioannidis JP, Loupy A. Prognostic Biomarkers in Kidney Transplantation: A Systematic Review and Critical Appraisal. J Am Soc Nephrol 2024; 35:177-188. [PMID: 38053242 PMCID: PMC10843205 DOI: 10.1681/asn.0000000000000260] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/08/2023] [Indexed: 12/07/2023] Open
Abstract
SIGNIFICANCE STATEMENT Why are there so few biomarkers accepted by health authorities and implemented in clinical practice, despite the high and growing number of biomaker studies in medical research ? In this meta-epidemiological study, including 804 studies that were critically appraised by expert reviewers, the authors have identified all prognostic kidney transplant biomarkers and showed overall suboptimal study designs, methods, results, interpretation, reproducible research standards, and transparency. The authors also demonstrated for the first time that the limited number of studies challenged the added value of their candidate biomarkers against standard-of-care routine patient monitoring parameters. Most biomarker studies tended to be single-center, retrospective studies with a small number of patients and clinical events. Less than 5% of the studies performed an external validation. The authors also showed the poor transparency reporting and identified a data beautification phenomenon. These findings suggest that there is much wasted research effort in transplant biomarker medical research and highlight the need to produce more rigorous studies so that more biomarkers may be validated and successfully implemented in clinical practice. BACKGROUND Despite the increasing number of biomarker studies published in the transplant literature over the past 20 years, demonstrations of their clinical benefit and their implementation in routine clinical practice are lacking. We hypothesized that suboptimal design, data, methodology, and reporting might contribute to this phenomenon. METHODS We formed a consortium of experts in systematic reviews, nephrologists, methodologists, and epidemiologists. A systematic literature search was performed in PubMed, Embase, Scopus, Web of Science, and Cochrane Library between January 1, 2005, and November 12, 2022 (PROSPERO ID: CRD42020154747). All English language, original studies investigating the association between a biomarker and kidney allograft outcome were included. The final set of publications was assessed by expert reviewers. After data collection, two independent reviewers randomly evaluated the inconsistencies for 30% of the references for each reviewer. If more than 5% of inconsistencies were observed for one given reviewer, a re-evaluation was conducted for all the references of the reviewer. The biomarkers were categorized according to their type and the biological milieu from which they were measured. The study characteristics related to the design, methods, results, and their interpretation were assessed, as well as reproducible research practices and transparency indicators. RESULTS A total of 7372 publications were screened and 804 studies met the inclusion criteria. A total of 1143 biomarkers were assessed among the included studies from blood ( n =821, 71.8%), intragraft ( n =169, 14.8%), or urine ( n =81, 7.1%) compartments. The number of studies significantly increased, with a median, yearly number of 31.5 studies (interquartile range [IQR], 23.8-35.5) between 2005 and 2012 and 57.5 (IQR, 53.3-59.8) between 2013 and 2022 ( P < 0.001). A total of 655 studies (81.5%) were retrospective, while 595 (74.0%) used data from a single center. The median number of patients included was 232 (IQR, 96-629) with a median follow-up post-transplant of 4.8 years (IQR, 3.0-6.2). Only 4.7% of studies were externally validated. A total of 346 studies (43.0%) did not adjust their biomarker for key prognostic factors, while only 3.1% of studies adjusted the biomarker for standard-of-care patient monitoring factors. Data sharing, code sharing, and registration occurred in 8.8%, 1.1%, and 4.6% of studies, respectively. A total of 158 studies (20.0%) emphasized the clinical relevance of the biomarker, despite the reported nonsignificant association of the biomarker with the outcome measure. A total of 288 studies assessed rejection as an outcome. We showed that these rejection studies shared the same characteristics as other studies. CONCLUSIONS Biomarker studies in kidney transplantation lack validation, rigorous design and methodology, accurate interpretation, and transparency. Higher standards are needed in biomarker research to prove the clinical utility and support clinical use.
Collapse
Affiliation(s)
- Marc Raynaud
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Solaf Al-Awadhi
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Kevin Louis
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Huanxi Zhang
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaojun Su
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Valentin Goutaudier
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Jiali Wang
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zeynep Demir
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Yongcheng Wei
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Agathe Truchot
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Antoine Bouquegneau
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liège, Liège, Belgium
| | - Arnaud Del Bello
- Department of Nephrology and Organ Transplantation, INSERM, CHU Rangueil & Purpan, Université Paul Sabatier, Toulouse, France
| | - Élodie Bailly
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yannis Lombardi
- Kidney Transplant Department, Tenon Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Mehdi Maanaoui
- Nephrology Department, CHU Lille, Lille University, Lille, France
- INSERM U1190, Translational Research for Diabetes, Lille, France
| | - Alessia Giarraputo
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Sofia Naser
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Gillian Divard
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Olivier Aubert
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | | | - Changxi Wang
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Longshan Liu
- The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Oriol Bestard
- Nephrology Department, Hospital de Vall d'Hebron, Barcelona, Spain
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - John J. Friedewald
- Division of Transplantation, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Leonardo Riella
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gary Collins
- Center for Statistics in Medicine, NDORMS, Botnar Research Center, University of Oxford, Oxford, United Kingdom
| | - John P.A. Ioannidis
- Departments of Medicine, of Epidemiology and Population Health, of Biomedical Data Science, and of Statistics and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California
| | - Alexandre Loupy
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| |
Collapse
|
11
|
Cleary SJ, Seo Y, Tian JJ, Kwaan N, Bulkley DP, Bentlage AEH, Vidarsson G, Boilard É, Spirig R, Zimring JC, Looney MR. IgG hexamers initiate acute lung injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577129. [PMID: 38328049 PMCID: PMC10849723 DOI: 10.1101/2024.01.24.577129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Antibodies can initiate lung injury in a variety of disease states such as autoimmunity, transfusion reactions, or after organ transplantation, but the key factors determining in vivo pathogenicity of injury-inducing antibodies are unclear. A previously overlooked step in complement activation by IgG antibodies has been elucidated involving interactions between IgG Fc domains that enable assembly of IgG hexamers, which can optimally activate the complement cascade. Here, we tested the in vivo relevance of IgG hexamers in a complement-dependent alloantibody model of acute lung injury. We used three approaches to block alloantibody hexamerization (antibody carbamylation, the K439E Fc mutation, or treatment with domain B from Staphylococcal protein A), all of which reduced acute lung injury. Conversely, Fc mutations promoting spontaneous hexamerization made a harmful alloantibody into a more potent inducer of acute lung injury and rendered an innocuous alloantibody pathogenic. Treatment with a recombinant Fc hexamer 'decoy' therapeutic protected mice from lung injury, including in a model with transgenic human FCGR2A expression that exacerbated pathology. These results indicate a direct in vivo role of IgG hexamerization in initiating acute lung injury and the potential for therapeutics that inhibit or mimic hexamerization to treat antibody-mediated diseases.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Yurim Seo
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Jennifer J. Tian
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - Nicholas Kwaan
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| | - David P. Bulkley
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF), CA, USA
| | | | | | - Éric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Rolf Spirig
- CSL Behring, Research, CSL Behring Biologics Research Center, Bern, Switzerland
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco (UCSF), CA, USA
| |
Collapse
|
12
|
Marco I, López-Azor García JC, González Martín J, Severo Sánchez A, García-Cosío Carmena MD, Mancebo Sierra E, de Juan Bagudá J, Castrodeza Calvo J, Hernández Pérez FJ, Delgado JF. De Novo Donor-Specific Antibodies after Heart Transplantation: A Comprehensive Guide for Clinicians. J Clin Med 2023; 12:7474. [PMID: 38068526 PMCID: PMC10707043 DOI: 10.3390/jcm12237474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 04/12/2024] Open
Abstract
Antibodies directed against donor-specific human leukocyte antigens (HLAs) can be detected de novo after heart transplantation and play a key role in long-term survival. De novo donor-specific antibodies (dnDSAs) have been associated with cardiac allograft vasculopathy, antibody-mediated rejection, and mortality. Advances in detection methods and international guideline recommendations have encouraged the adoption of screening protocols among heart transplant units. However, there is still a lack of consensus about the correct course of action after dnDSA detection. Treatment is usually started when antibody-mediated rejection is present; however, some dnDSAs appear years before graft failure is detected, and at this point, damage may be irreversible. In particular, class II, anti-HLA-DQ, complement binding, and persistent dnDSAs have been associated with worse outcomes. Growing evidence points towards a more aggressive management of dnDSA. For that purpose, better diagnostic tools are needed in order to identify subclinical graft injury. Cardiac magnetic resonance, strain techniques, or coronary physiology parameters could provide valuable information to identify patients at risk. Treatment of dnDSA usually involves plasmapheresis, intravenous immunoglobulin, immunoadsorption, and ritxumab, but the benefit of these therapies is still controversial. Future efforts should focus on establishing effective treatment protocols in order to improve long-term survival of heart transplant recipients.
Collapse
Affiliation(s)
- Irene Marco
- Cardiology Department, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | - Juan Carlos López-Azor García
- Cardiology Department, Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain; (J.C.L.-A.G.); (F.J.H.P.)
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.G.M.); (M.D.G.-C.C.); (J.d.J.B.); (J.C.C.)
- School of Medicine, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Javier González Martín
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.G.M.); (M.D.G.-C.C.); (J.d.J.B.); (J.C.C.)
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Andrea Severo Sánchez
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - María Dolores García-Cosío Carmena
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.G.M.); (M.D.G.-C.C.); (J.d.J.B.); (J.C.C.)
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Esther Mancebo Sierra
- Immunology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Javier de Juan Bagudá
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.G.M.); (M.D.G.-C.C.); (J.d.J.B.); (J.C.C.)
- School of Medicine, Universidad Europea de Madrid, 28670 Madrid, Spain
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Javier Castrodeza Calvo
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.G.M.); (M.D.G.-C.C.); (J.d.J.B.); (J.C.C.)
- Cardiology Department, Hospital Universitario Gregorio Marañón, 28007 Madrid, Spain
| | | | - Juan Francisco Delgado
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), 28029 Madrid, Spain; (J.G.M.); (M.D.G.-C.C.); (J.d.J.B.); (J.C.C.)
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
- School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
13
|
Al-Awadhi S, Raynaud M, Louis K, Bouquegneau A, Taupin JL, Aubert O, Loupy A, Lefaucheur C. Complement-activating donor-specific anti-HLA antibodies in solid organ transplantation: systematic review, meta-analysis, and critical appraisal. Front Immunol 2023; 14:1265796. [PMID: 37849755 PMCID: PMC10577173 DOI: 10.3389/fimmu.2023.1265796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/07/2023] [Indexed: 10/19/2023] Open
Abstract
Introduction Several studies have investigated the impact of circulating complement-activating anti-human leukocyte antigen donor-specific antibodies (anti-HLA DSAs) on organ transplant outcomes. However, a critical appraisal of these studies and a demonstration of the prognostic value of complement-activating status over anti-HLA DSA mean fluorescence intensity (MFI) level are lacking. Methods We conducted a systematic review, meta-analysis and critical appraisal evaluating the role of complement-activating anti-HLA DSAs on allograft outcomes in different solid organ transplants. We included studies through Medline, Cochrane, Scopus, and Embase since inception of databases till May 05, 2023. We evaluated allograft loss as the primary outcome, and allograft rejection as the secondary outcome. We used the Newcastle-Ottawa Scale and funnel plots to assess risk of bias and used bias adjustment methods when appropriate. We performed multiple subgroup analyses to account for sources of heterogeneity and studied the added value of complement assays over anti-HLA DSA MFI level. Results In total, 52 studies were included in the final meta-analysis (11,035 patients). Complement-activating anti-HLA DSAs were associated with an increased risk of allograft loss (HR 2.77; 95% CI 2.33-3.29, p<0.001; I²=46.2%), and allograft rejection (HR 4.98; 95% CI 2.96-8.36, p<0.01; I²=70.9%). These results remained significant after adjustment for potential sources of bias and across multiple subgroup analyses. After adjusting on pan-IgG anti-HLA DSA defined by the MFI levels, complement-activating anti-HLA DSAs were significantly and independently associated with an increased risk of allograft loss. Discussion We demonstrated in this systematic review, meta-analysis and critical appraisal the significant deleterious impact and the independent prognostic value of circulating complement-activating anti-HLA DSAs on solid organ transplant risk of allograft loss and rejection.
Collapse
Affiliation(s)
- Solaf Al-Awadhi
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Marc Raynaud
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Kevin Louis
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Antoine Bouquegneau
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
- Department of Nephrology, Dialysis and Transplantation, Centre Hospitalier Universitaire (CHU) de Liège, Liège, Belgium
| | - Jean-Luc Taupin
- Department of Immunology and Histocompatibility, Centre Hospitalier Universitaire (CHU) Paris–GH St–Louis Lariboisière, Paris, France
| | - Olivier Aubert
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Alexandre Loupy
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Carmen Lefaucheur
- Université de Paris Cité, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche (UMR)-S970, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Centre for Organ Transplantation, Paris, France
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
14
|
Townsend M, Pidborochynski T, Cantor RS, Khoury M, Campbell P, Halpin A, Urschel S, Kim D, Nahirniak S, West LJ, Buchholz H, Conway J. Prospective examination of HLA sensitization after VAD implantation in children and adults. Transpl Immunol 2023; 80:101892. [PMID: 37419373 DOI: 10.1016/j.trim.2023.101892] [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: 05/12/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023]
Abstract
BACKGROUND Ventricular assist devices (VADs) have improved survival to heart transplantation (HTx). However, VADs have been associated with development of antibodies against human leukocyte antigen (HLA-Ab) which may limit the donor pool and decrease survival post-HTx. Since HLA-Ab development after VAD insertion is poorly understood, the purpose of this prospective single-center study was to quantify the incidence of and evaluate risk factors for HLA-Ab development across the age spectrum following VAD implantation. METHODS Adult and pediatric patients undergoing VAD placement as bridge to transplant or transplant candidacy between 5/2016 and 7/2020 were enrolled. HLA-Ab were assessed pre-VAD and at 1-, 3-, and 12-months post-implant. Factors associated with HLA-Ab development post-VAD implant were explored using univariate and multivariate logistic regression. RESULTS 15/41 (37%) adults and 7/17 (41%) children developed new HLA-Ab post-VAD. The majority of patients (19/22) developed HLA-Ab within two months of implant. New class I HLA-Ab were more common (87% adult, 86% pediatric). Prior pregnancy was strongly associated with HLA-Ab development in adults post-VAD (HR 16.7, 95% CI 1.8-158, p = 0.01). Of the patients who developed new HLA-Ab post-VAD, in 45% (10/22) the HLA-Ab resolved while in 55% (12/22) the HLA-Ab persisted. CONCLUSION More than one-third of adult and pediatric VAD patients developed new HLA-Ab early after VAD implant with the majority having class I antibodies. Prior pregnancy was strongly associated with post-VAD HLA-Ab development. Further studies are needed to predict regression or persistence of HLA-Ab developed post-VAD, to understand modulation of individuals' immune responses to sensitizing events, and to determine whether transiently detected HLA-Ab post-VAD recur and have long-term clinical impact post-heart transplantation.
Collapse
Affiliation(s)
- Madeleine Townsend
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada.
| | - Tara Pidborochynski
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada
| | - Ryan S Cantor
- Kirklin Solutions, Birmingham, AL, United States of America
| | - Michael Khoury
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada; Canada Donation And Transplantation Research Program, University of Alberta, Edmonton, AB, Canada
| | - Patricia Campbell
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada; Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Anne Halpin
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada; Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Simon Urschel
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada; Canada Donation And Transplantation Research Program, University of Alberta, Edmonton, AB, Canada; Department of Surgery, University of Alberta, Edmonton, AB, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Daniel Kim
- Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada; Department of Medicine, University of Alberta, Edmonton, AB, Canada; Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Susan Nahirniak
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Lori J West
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada; Canada Donation And Transplantation Research Program, University of Alberta, Edmonton, AB, Canada; Department of Surgery, University of Alberta, Edmonton, AB, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Holger Buchholz
- Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada; Department of Medicine, University of Alberta, Edmonton, AB, Canada; Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
| | - Jennifer Conway
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
van den Broek DAJ, Meziyerh S, Budde K, Lefaucheur C, Cozzi E, Bertrand D, López del Moral C, Dorling A, Emonds MP, Naesens M, de Vries APJ. The Clinical Utility of Post-Transplant Monitoring of Donor-Specific Antibodies in Stable Renal Transplant Recipients: A Consensus Report With Guideline Statements for Clinical Practice. Transpl Int 2023; 36:11321. [PMID: 37560072 PMCID: PMC10408721 DOI: 10.3389/ti.2023.11321] [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: 03/01/2023] [Accepted: 06/22/2023] [Indexed: 08/11/2023]
Abstract
Solid phase immunoassays improved the detection and determination of the antigen-specificity of donor-specific antibodies (DSA) to human leukocyte antigens (HLA). The widespread use of SPI in kidney transplantation also introduced new clinical dilemmas, such as whether patients should be monitored for DSA pre- or post-transplantation. Pretransplant screening through SPI has become standard practice and DSA are readily determined in case of suspected rejection. However, DSA monitoring in recipients with stable graft function has not been universally established as standard of care. This may be related to uncertainty regarding the clinical utility of DSA monitoring as a screening tool. This consensus report aims to appraise the clinical utility of DSA monitoring in recipients without overt signs of graft dysfunction, using the Wilson & Junger criteria for assessing the validity of a screening practice. To assess the evidence on DSA monitoring, the European Society for Organ Transplantation (ESOT) convened a dedicated workgroup, comprised of experts in transplantation nephrology and immunology, to review relevant literature. Guidelines and statements were developed during a consensus conference by Delphi methodology that took place in person in November 2022 in Prague. The findings and recommendations of the workgroup on subclinical DSA monitoring are presented in this article.
Collapse
Affiliation(s)
- Dennis A. J. van den Broek
- Division of Nephrology, Department of Medicine, Leiden Transplant Center, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Soufian Meziyerh
- Division of Nephrology, Department of Medicine, Leiden Transplant Center, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, Kidney Transplant Department, Saint Louis Hospital, Université de Paris Cité, Paris, France
| | - Emanuele Cozzi
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, Transplant Immunology Unit, Padua University Hospital, Padua, Italy
| | - Dominique Bertrand
- Department of Nephrology, Transplantation and Hemodialysis, Rouen University Hospital, Rouen, France
| | - Covadonga López del Moral
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Valdecilla Biomedical Research Institute (IDIVAL), Santander, Spain
| | - Anthony Dorling
- Department of Inflammation Biology, Centre for Nephrology, Urology and Transplantation, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Marie-Paule Emonds
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross-Flanders, Mechelen, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Aiko P. J. de Vries
- Division of Nephrology, Department of Medicine, Leiden Transplant Center, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | | |
Collapse
|
16
|
Couzi L, Malvezzi P, Amrouche L, Anglicheau D, Blancho G, Caillard S, Freist M, Guidicelli GL, Kamar N, Lefaucheur C, Mariat C, Koenig A, Noble J, Thaunat O, Thierry A, Taupin JL, Bertrand D. Imlifidase for Kidney Transplantation of Highly Sensitized Patients With a Positive Crossmatch: The French Consensus Guidelines. Transpl Int 2023; 36:11244. [PMID: 37448448 PMCID: PMC10336835 DOI: 10.3389/ti.2023.11244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023]
Abstract
Imlifidase recently received early access authorization for highly sensitized adult kidney transplant candidates with a positive crossmatch against an ABO-compatible deceased donor. These French consensus guidelines have been generated by an expert working group, in order to homogenize patient selection, associated treatments and follow-up. This initiative is part of an international effort to analyze properly the benefits and tolerance of this new costly treatment in real-life. Eligible patients must meet the following screening criteria: cPRA ≥ 98%, ≤ 65-year of age, ≥ 3 years on the waiting list, and a low risk of biopsy-related complications. The final decision to use Imlifidase will be based on the two following criteria. First, the results of a virtual crossmatch on recent serum, which shall show a MFI for the immunodominant donor-specific antibodies (DSA) > 6,000 but the value of which does not exceed 5,000 after 1:10 dilution. Second, the post-Imlifidase complement-dependent cytotoxicity crossmatch must be negative. Patients treated with Imlifidase will receive an immunosuppressive regimen based on steroids, rATG, high dose IVIg, rituximab, tacrolimus and mycophenolic acid. Frequent post-transplant testing for DSA and systematic surveillance kidney biopsies are highly recommended to monitor post-transplant DSA rebound and subclinical rejection.
Collapse
Affiliation(s)
- Lionel Couzi
- Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
- CNRS-UMR 5164 Immuno ConcEpT, Université de Bordeaux, Bordeaux, France
| | - Paolo Malvezzi
- Centre Hospitalier Universitaire de Grenoble, La Tronche, France
| | | | | | - Gilles Blancho
- Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | | | - Marine Freist
- Centre Hospitalier Emile Roux, Le Puy-en-Velay, France
| | | | - Nassim Kamar
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Christophe Mariat
- Centre Hospitalier Universitaire (CHU) de Saint-Étienne, Saint-Etienne, France
| | | | - Johan Noble
- Centre Hospitalier Universitaire de Grenoble, La Tronche, France
| | | | - Antoine Thierry
- Centre Hospitalier Universitaire (CHU) de Poitiers, Poitiers, France
| | | | | |
Collapse
|
17
|
Gibson B, Connelly C, Moldakhmetova S, Sheerin NS. Complement activation and kidney transplantation; a complex relationship. Immunobiology 2023; 228:152396. [PMID: 37276614 DOI: 10.1016/j.imbio.2023.152396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/07/2023]
Abstract
Although kidney transplantation is the best treatment for end stage kidney disease, the benefits are limited by factors such as the short fall in donor numbers, the burden of immunosuppression and graft failure. Although there have been improvements in one-year outcomes, the annual rate of graft loss beyond the first year has not significantly improved, despite better therapies to control the alloimmune response. There is therefore a need to develop alternative strategies to limit kidney injury at all stages along the transplant pathway and so improve graft survival. Complement is primarily part of the innate immune system, but is also known to enhance the adaptive immune response. There is increasing evidence that complement activation occurs at many stages during transplantation and can have deleterious effects on graft outcome. Complement activation begins in the donor and occurs again on reperfusion following a period of ischemia. Complement can contribute to the development of the alloimmune response and may directly contribute to graft injury during acute and chronic allograft rejection. The complexity of the relationship between complement activation and allograft outcome is further increased by the capacity of the allograft to synthesise complement proteins, the contribution complement makes to interstitial fibrosis and complement's role in the development of recurrent disease. The better we understand the role played by complement in kidney transplant pathology the better placed we will be to intervene. This is particularly relevant with the rapid development of complement therapeutics which can now target different the different pathways of the complement system. Combining our basic understanding of complement biology with preclinical and observational data will allow the development and delivery of clinical trials which have best chance to identify any benefit of complement inhibition.
Collapse
Affiliation(s)
- B Gibson
- Clinical and Translational Research Institute Faculty of Medical Sciences, Newcastle University Newcastle upon Tyne, NE2 4HH, UK
| | - C Connelly
- Clinical and Translational Research Institute Faculty of Medical Sciences, Newcastle University Newcastle upon Tyne, NE2 4HH, UK
| | - S Moldakhmetova
- Clinical and Translational Research Institute Faculty of Medical Sciences, Newcastle University Newcastle upon Tyne, NE2 4HH, UK
| | - N S Sheerin
- Clinical and Translational Research Institute Faculty of Medical Sciences, Newcastle University Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
18
|
Zhang Y, He J, Yang Z, Zheng H, Deng H, Luo Z, Sun Q, Sun Q. Preventative effect of TSPO ligands on mixed antibody-mediated rejection through a Mitochondria-mediated metabolic disorder. J Transl Med 2023; 21:295. [PMID: 37131248 PMCID: PMC10152746 DOI: 10.1186/s12967-023-04134-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/13/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Immune-mediated rejection was the major cause of graft dysfunction. Although the advances in immunosuppressive agents have markedly reduced the incidence of T-cell-mediated rejection after transplantation. However, the incidence of antibody-mediated rejection (AMR) remains high. Donor-specific antibodies (DSAs) were considered the major mediators of allograft loss. Previously, we showed that treatment with 18-kDa translocator protein (TSPO) ligands inhibited the differentiation and effector functions of T cells and reduced the rejection observed after allogeneic skin transplantation in mice. This study we further investigate the effect of TSPO ligands on B cells and DSAs production in the recipients of mixed-AMR model. METHODS In vitro, we explored the effect of treatment with TSPO ligands on the activation, proliferation, and antibody production of B cells. Further, we established a heart-transplantation mixed-AMR model in rats. This model was treated with the TSPO ligands, FGIN1-27 or Ro5-4864, to investigate the role of ligands in preventing transplant rejection and DSAs production in vivo. As TSPO was the mitochondrial membrane transporters, we then investigated the TSPO ligands effect on mitochondrial-related metabolic ability of B cells as well as expression of downstream proteins. RESULTS In vitro studies, treatment with TSPO ligands inhibited the differentiation of B cells into CD138+CD27+ plasma cells; reduced antibodies, IgG and IgM, secretion of B cells; and suppressed the B cell activation and proliferation. In the mixed-AMR rat model, treatment with FGIN1-27 or Ro5-4864 attenuated DSA-mediated cardiac-allograft injury, prolonged graft survival, and reduced the numbers of B cells, including IgG+ secreting B cells, T cells and macrophages infiltrating in grafts. For the further mechanism exploration, treatment with TSPO ligands inhibited the metabolic ability of B cells by downregulating expression of pyruvate dehydrogenase kinase 1 and proteins in complexes I, II, and IV of the electron transport chain. CONCLUSIONS We clarified the mechanism of action of TSPO ligands on B-cell functions and provided new ideas and drug targets for the clinical treatment of postoperative AMR.
Collapse
Affiliation(s)
- Yannan Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiannan He
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhe Yang
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Haofeng Zheng
- Division of kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 2nd road Zhongshan, Yuexiu District, Guangzhou, 510080, China
| | - Haoxiang Deng
- Division of kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 2nd road Zhongshan, Yuexiu District, Guangzhou, 510080, China
| | - Zihuan Luo
- Division of kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 2nd road Zhongshan, Yuexiu District, Guangzhou, 510080, China
| | - Qipeng Sun
- Division of kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 2nd road Zhongshan, Yuexiu District, Guangzhou, 510080, China
| | - Qiquan Sun
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Division of kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 2nd road Zhongshan, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
19
|
Wong ZE, Downing J, De Santis D, Halse M, Bruce S, Truong L, Martinez P, D'Orsogna LJ. C3d-binding assay for the detection of complement activating HLA antibodies: A useful tool for allocation to highly sensitised recipients in the post-CDC era? HLA 2023. [PMID: 36851856 DOI: 10.1111/tan.15010] [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: 08/31/2022] [Revised: 01/23/2023] [Accepted: 02/23/2023] [Indexed: 03/01/2023]
Abstract
The CDC crossmatch test is being phased out in solid organ donor allocation, and standard luminex single antigen bead assays do not differentiate complement activating function of HLA antibodies. The current study investigated the LIFECODES C3d-binding assay to determine if it could accurately predict actual T and B cell CDC results in a cohort of highly sensitised patients. Nineteen serum samples from different highly sensitised solid organ patients were crossmatched against cells from 62 unique donors, with 174 total T and B cell crossmatches performed. The sera also underwent SAB assay using OLI and LC platforms, and C3d-binding assay. Complement activating ability of each unique HLA antibody specificity detected using SAB was assigned based on the actual CDC results, which was then used to determine the accuracy of the C3d-binding assay. The C3d-binding assay was found to be highly accurate, with sensitivity of 95%, specificity 89% and negative predictive value 97% for class I DSA and the T cell CDC crossmatch results. Furthermore, we found 100% accuracy for prediction of the complement activating function of HLA-C antibodies. Negative predictive value of above 90% was also found for HLA class II DSA. C3d-binding proved more accurate than virtual crossmatch alone to predict CDC results. This study confirms that the C3d-binding assay predicts actual CDC crossmatch results accurately. In particular, the high negative predictive value of the C3d-binding assay may be extremely useful to define HLA antibodies that do not activate complement in highly sensitised recipients.
Collapse
Affiliation(s)
- Zo Ee Wong
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Jonathan Downing
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Dianne De Santis
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Megan Halse
- School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Samuel Bruce
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Linh Truong
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,School of Biomedical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Patricia Martinez
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Lloyd J D'Orsogna
- Department of Clinical Immunology and PathWest, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
20
|
Stringer D, Gardner L, Shaw O, Clarke B, Briggs D, Worthington J, Buckland M, Danzi G, Hilton R, Picton M, Thuraisingham R, Borrows R, Baker R, McCullough K, Stoves J, Phanish M, Shah S, Shiu KY, Walsh SB, Ahmed A, Ayub W, Hegarty J, Tinch-Taylor R, Georgiou E, Bidad N, Kılıç A, Moon Z, Horne R, McCrone P, Kelly J, Murphy C, Peacock J, Dorling A. Optimized immunosuppression to prevent graft failure in renal transplant recipients with HLA antibodies (OuTSMART): a randomised controlled trial. EClinicalMedicine 2023; 56:101819. [PMID: 36684392 PMCID: PMC9852275 DOI: 10.1016/j.eclinm.2022.101819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND 3% of kidney transplant recipients return to dialysis annually upon allograft failure. Development of antibodies (Ab) against human leukocyte antigens (HLA) is a validated prognostic biomarker of allograft failure. We tested whether screening for HLA Ab, combined with an intervention to improve adherence and optimization of immunosuppression could prevent allograft failure. METHODS Prospective, open-labelled randomised biomarker-based strategy (hybrid) trial in 13 UK transplant centres [EudraCT (2012-004308-36) and ISRCTN (46157828)]. Patients were randomly allocated (1:1) to unblinded or double-blinded arms and screened every 8 months. Unblinded HLA Ab+ patients were interviewed to encourage medication adherence and had tailored optimisation of Tacrolimus, Mycophenolate mofetil and Prednisolone. The primary outcome was time to graft failure in an intention to treat analysis. The trial had 80% power to detect a hazard ratio of 0.49 in donor specific antibody (DSA)+ patients. FINDINGS From 11/9/13 to 27/10/16, 5519 were screened for eligibility and 2037 randomised (1028 to unblinded care and 1009 to double blinded care). We identified 198 with DSA and 818 with non-DSA. Development of DSA, but not non-DSA was predictive of graft failure. HRs for graft failure in unblinded DSA+ and non-DSA+ groups were 1.54 (95% CI: 0.72 to 3.30) and 0.97 (0.54-1.74) respectively, providing no evidence of an intervention effect. Non-inferiority for the overall unblinded versus blinded comparison was not demonstrated as the upper confidence limit of the HR for graft failure exceeded 1.4 (1.02, 95% CI: 0.72 to 1.44). The only secondary endpoint reduced in the unblinded arm was biopsy-proven rejection. INTERPRETATION Intervention to improve adherence and optimize immunosuppression does not delay failure of renal transplants after development of DSA. Whilst DSA predicts increased risk of allograft failure, novel interventions are needed before screening can be used to direct therapy. FUNDING The National Institute for Health Research Efficacy and Mechanism Evaluation programme grant (ref 11/100/34).
Collapse
Affiliation(s)
- Dominic Stringer
- Biostatistics and Health Informatics, The Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- King's Clinical Trials Unit, King's College London, London, UK
| | - Leanne Gardner
- King's Clinical Trials Unit, King's College London, London, UK
- Centre for Nephrology, Urology and Transplantation, Department of Inflammation Biology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Olivia Shaw
- Clinical Transplantation Laboratory, Viapath Analytics LLP, London, UK
| | - Brendan Clarke
- Transplant Immunology, Level 09 Gledhow Wing, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - David Briggs
- NHSBT Birmingham, Vincent Drive, Edgbaston, Birmingham, B15 2SG, UK
| | - Judith Worthington
- Transplantation Laboratory, Manchester Royal Infirmary, Oxford Road, Manchester, M13 9WL, UK
| | - Matthew Buckland
- Clinical Transplantation Laboratory, The Royal London Hospital, 2nd Floor, Pathology and Pharmacy Building, 80 Newark Street, London, E1 1BB, UK
| | - Guilherme Danzi
- Renal Unit, Hospital das Clínicas da Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife - PE, 50670-901, Brazil
| | - Rachel Hilton
- Department of Nephrology and Transplantation, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Michael Picton
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Road, Manchester, M13 9WL, UK
| | - Raj Thuraisingham
- Department of Renal Medicine and Transplantation, Barts Health NHS Trust, London, E1 1BB, UK
| | - Richard Borrows
- Renal Unit, University Hospital Birmingham, Edgbaston, Birmingham, B15 2LN, UK
| | - Richard Baker
- Renal Unit, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | - Keith McCullough
- Renal Unit, York Teaching Hospital NHS Foundation Trust, York, YO31 8HE, UK
| | - John Stoves
- Renal Unit, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, BD5 0NA, UK
| | - Mysore Phanish
- Renal Unit, Epsom and St Helier University Hospitals NHS Trust, Surrey, UK
| | - Sapna Shah
- Renal Unit, King's College Hospital, London, SE5 9RJ, UK
| | - Kin Yee Shiu
- UCL Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, NW3 2QG, UK
| | - Stephen B. Walsh
- UCL Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, NW3 2QG, UK
| | - Aimun Ahmed
- Renal Unit, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, PR2 9HT, UK
| | - Waqar Ayub
- Renal Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Janet Hegarty
- Renal Unit, Salford Royal NHS Foundation Trust, Salford, M6 8HD, UK
| | - Rose Tinch-Taylor
- Biostatistics and Health Informatics, The Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- King's Clinical Trials Unit, King's College London, London, UK
| | | | - Natalie Bidad
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Ayşenur Kılıç
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Zoe Moon
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Robert Horne
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, WC1H 9JP, UK
| | - Paul McCrone
- King's Clinical Trials Unit, King's College London, London, UK
- Faculty of Education, Health and Human Sciences, University of Greenwich, London, UK
| | - Joanna Kelly
- King's Clinical Trials Unit, King's College London, London, UK
| | - Caroline Murphy
- King's Clinical Trials Unit, King's College London, London, UK
| | - Janet Peacock
- School of Life Course and Population Sciences, King's College London, London, UK
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth College, USA
| | - Anthony Dorling
- Centre for Nephrology, Urology and Transplantation, Department of Inflammation Biology, King's College London, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
- Corresponding author.
| |
Collapse
|
21
|
Gaykema LH, van Nieuwland RY, Dekkers MC, van Essen MF, Heidt S, Zaldumbide A, van den Berg CW, Rabelink TJ, van Kooten C. Inhibition of complement activation by CD55 overexpression in human induced pluripotent stem cell derived kidney organoids. Front Immunol 2023; 13:1058763. [PMID: 36713440 PMCID: PMC9880527 DOI: 10.3389/fimmu.2022.1058763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023] Open
Abstract
End stage renal disease is an increasing problem worldwide driven by aging of the population and increased prevalence of metabolic disorders and cardiovascular disease. Currently, kidney transplantation is the only curative option, but donor organ shortages greatly limit its application. Regenerative medicine has the potential to solve the shortage by using stem cells to grow the desired tissues, like kidney tissue. Immune rejection poses a great threat towards the implementation of stem cell derived tissues and various strategies have been explored to limit the immune response towards these tissues. However, these studies are limited by targeting mainly T cell mediated immune rejection while the rejection process also involves innate and humoral immunity. In this study we investigate whether inhibition of the complement system in human induced pluripotent stem cells (iPSC) could provide protection from such immune injury. To this end we created knock-in iPSC lines of the membrane bound complement inhibitor CD55 to create a transplant-specific protection towards complement activation. CD55 inhibits the central driver of the complement cascade, C3 convertase, and we show that overexpression is able to decrease complement activation on both iPSCs as well as differentiated kidney organoids upon stimulation with anti-HLA antibodies to mimic the mechanism of humoral rejection.
Collapse
Affiliation(s)
- Lonneke H. Gaykema
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Rianne Y. van Nieuwland
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Mette C. Dekkers
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Mieke F. van Essen
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands,Eurotransplant Reference Laboratory, Leiden University Medical Center, Leiden, Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Cathelijne W. van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands,The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands,The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Ton J. Rabelink,
| | - Cees van Kooten
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
22
|
Seeking Standardized Definitions for HLA-incompatible Kidney Transplants: A Systematic Review. Transplantation 2023; 107:231-253. [PMID: 35915547 DOI: 10.1097/tp.0000000000004262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is no standard definition for "HLA incompatible" transplants. For the first time, we systematically assessed how HLA incompatibility was defined in contemporary peer-reviewed publications and its prognostic implication to transplant outcomes. METHODS We combined 2 independent searches of MEDLINE, EMBASE, and the Cochrane Library from 2015 to 2019. Content-expert reviewers screened for original research on outcomes of HLA-incompatible transplants (defined as allele or molecular mismatch and solid-phase or cell-based assays). We ascertained the completeness of reporting on a predefined set of variables assessing HLA incompatibility, therapies, and outcomes. Given significant heterogeneity, we conducted narrative synthesis and assessed risk of bias in studies examining the association between death-censored graft failure and HLA incompatibility. RESULTS Of 6656 screened articles, 163 evaluated transplant outcomes by HLA incompatibility. Most articles reported on cytotoxic/flow T-cell crossmatches (n = 98). Molecular genotypes were reported for selected loci at the allele-group level. Sixteen articles reported on epitope compatibility. Pretransplant donor-specific HLA antibodies were often considered (n = 143); yet there was heterogeneity in sample handling, assay procedure, and incomplete reporting on donor-specific HLA antibodies assignment. Induction (n = 129) and maintenance immunosuppression (n = 140) were frequently mentioned but less so rejection treatment (n = 72) and desensitization (n = 70). Studies assessing death-censored graft failure risk by HLA incompatibility were vulnerable to bias in the participant, predictor, and analysis domains. CONCLUSIONS Optimization of transplant outcomes and personalized care depends on accurate HLA compatibility assessment. Reporting on a standard set of variables will help assess generalizability of research, allow knowledge synthesis, and facilitate international collaboration in clinical trials.
Collapse
|
23
|
Clinical recommendations for posttransplant assessment of anti-HLA (Human Leukocyte Antigen) donor-specific antibodies: A Sensitization in Transplantation: Assessment of Risk consensus document. Am J Transplant 2023; 23:115-132. [PMID: 36695614 DOI: 10.1016/j.ajt.2022.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 01/13/2023]
Abstract
Although anti-HLA (Human Leukocyte Antigen) donor-specific antibodies (DSAs) are commonly measured in clinical practice and their relationship with transplant outcome is well established, clinical recommendations for anti-HLA antibody assessment are sparse. Supported by a careful and critical review of the current literature performed by the Sensitization in Transplantation: Assessment of Risk 2022 working group, this consensus report provides clinical practice recommendations in kidney, heart, lung, and liver transplantation based on expert assessment of quality and strength of evidence. The recommendations address 3 major clinical problems in transplantation and include guidance regarding posttransplant DSA assessment and application to diagnostics, prognostics, and therapeutics: (1) the clinical implications of positive posttransplant DSA detection according to DSA status (ie, preformed or de novo), (2) the relevance of posttransplant DSA assessment for precision diagnosis of antibody-mediated rejection and for treatment management, and (3) the relevance of posttransplant DSA for allograft prognosis and risk stratification. This consensus report also highlights gaps in current knowledge and provides directions for clinical investigations and trials in the future that will further refine the clinical utility of posttransplant DSA assessment, leading to improved transplant management and patient care.
Collapse
|
24
|
Tambur AR, Bestard O, Campbell P, Chong AS, Barrio MC, Ford ML, Gebel HM, Heidt S, Hickey M, Jackson A, Kosmoliaptsis V, Lefaucheur C, Louis K, Mannon RB, Mengel M, Morris A, Pinelli DF, Reed EF, Schinstock C, Taupin JL, Valenzuela N, Wiebe C, Nickerson P. Sensitization in transplantation: Assessment of Risk 2022 Working Group Meeting Report. Am J Transplant 2023; 23:133-149. [PMID: 36695615 DOI: 10.1016/j.ajt.2022.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/20/2022] [Accepted: 11/02/2022] [Indexed: 01/13/2023]
Abstract
The Sensitization in Transplantation: Assessment of Risk workgroup is a collaborative effort of the American Society of Transplantation and the American Society of Histocompatibility and Immunogenetics that aims at providing recommendations for clinical testing, highlights gaps in current knowledge, and proposes areas for further research to enhance histocompatibility testing in support of solid organ transplantation. This report provides updates on topics discussed by the previous Sensitization in Transplantation: Assessment of Risk working groups and introduces 2 areas of exploration: non-human leukocyte antigen antibodies and utilization of human leukocyte antigen antibody testing measurement to evaluate the efficacy of antibody-removal therapies.
Collapse
Affiliation(s)
- Anat R Tambur
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA.
| | - Oriol Bestard
- Vall d'Hebron Institut de Recerca, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Patricia Campbell
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Anita S Chong
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Martha Crespo Barrio
- Department of Nephrology, Hospital del Mar & Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, Georgia, USA
| | - Howard M Gebel
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Netherlands
| | - Michelle Hickey
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Annette Jackson
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| | - Kevin Louis
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| | - Roslyn B Mannon
- Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Michael Mengel
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Anna Morris
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David F Pinelli
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Jean-Luc Taupin
- Department of Immunology, Saint Louis Hospital and University Paris-Cité, Paris, France
| | - Nicole Valenzuela
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter Nickerson
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
25
|
Jaiswal A, Bell J, DeFilippis EM, Kransdorf EP, Patel J, Kobashigawa JA, Kittleson MM, Baran DA. Assessment and management of allosensitization following heart transplant in adults. J Heart Lung Transplant 2022; 42:423-432. [PMID: 36702686 DOI: 10.1016/j.healun.2022.12.011] [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: 09/23/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Immunological injury to the allograft, specifically by antibodies to de novo donor specific human leukocyte antigen (dnDSA) and antibody mediated injury and rejection are the major limitations to graft survival after heart transplantation (HT). As such, our approach to allosensitization remains limited by the inability of contemporaneous immunoassays to unravel pathogenic potential of dnDSA. Additionally, the role of dnDSA is continuously evaluated with emerging methods to detect rejection. Moreover, the timing and frequency of dnDSA monitoring for early detection and risk mitigation as well as management of dnDSA remain challenging. A strategic approach to dnDSA employs diagnostic assays to determine relevant antibodies in conjunction with clinical presentation and injury/rejection of allograft to tailor therapeutics. In this review, we aim to outline contemporary knowledge involving detection, monitoring and management of dnDSA after HT. Subsequently, we propose a diagnostic and therapeutic approach that may mitigate morbidity and mortality while balancing adverse reactions from pharmacotherapy.
Collapse
Affiliation(s)
- Abhishek Jaiswal
- Hartford HealthCare Heart and Vascular Institute, Hartford Hospital, Hartford, Connecticut.
| | - Jennifer Bell
- Hartford HealthCare Heart and Vascular Institute, Hartford Hospital, Hartford, Connecticut
| | - Ersilia M DeFilippis
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, New York
| | - Evan P Kransdorf
- Division of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jignesh Patel
- Division of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jon A Kobashigawa
- Division of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michelle M Kittleson
- Division of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - David A Baran
- Cleveland Clinic, Heart, Vascular and Thoracic Institute, Advanced Heart Failure Program, Weston, Florida
| |
Collapse
|
26
|
Anwar IJ, DeLaura I, Ladowski J, Gao Q, Knechtle SJ, Kwun J. Complement-targeted therapies in kidney transplantation-insights from preclinical studies. Front Immunol 2022; 13:984090. [PMID: 36311730 PMCID: PMC9606228 DOI: 10.3389/fimmu.2022.984090] [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: 07/01/2022] [Accepted: 09/28/2022] [Indexed: 01/21/2023] Open
Abstract
Aberrant activation of the complement system contributes to solid-organ graft dysfunction and failure. In kidney transplantation, the complement system is implicated in the pathogenesis of antibody- and cell-mediated rejection, ischemia-reperfusion injury, and vascular injury. This has led to the evaluation of select complement inhibitors (e.g., C1 and C5 inhibitors) in clinical trials with mixed results. However, the complement system is highly complex: it is composed of more than 50 fluid-phase and surface-bound elements, including several complement-activated receptors-all potential therapeutic targets in kidney transplantation. Generation of targeted pharmaceuticals and use of gene editing tools have led to an improved understanding of the intricacies of the complement system in allo- and xeno-transplantation. This review summarizes our current knowledge of the role of the complement system as it relates to rejection in kidney transplantation, specifically reviewing evidence gained from pre-clinical models (rodent and nonhuman primate) that may potentially be translated to clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Stuart J. Knechtle
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Jean Kwun
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
27
|
Louis K, Lefaucheur C. DSA in solid organ transplantation: is it a matter of specificity, amount, or functional characteristics? Curr Opin Organ Transplant 2022; 27:392-398. [PMID: 35881421 DOI: 10.1097/mot.0000000000001006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The present review describes the clinical relevance of human leukocyte antigen (HLA) donor-specific antibodies (HLA-DSAs) as biomarkers of alloimmunity and summarizes recent improvements in their characterization that provide insights into immune risk assessment, precision diagnosis, and prognostication in transplantation. RECENT FINDINGS Recent studies have addressed the clinical utility of HLA-DSAs as biomarkers for immune risk assessment in pretransplant and peritransplant, diagnosis and treatment evaluation of antibody-mediated rejection, immune monitoring posttransplant, and risk stratification. SUMMARY HLA-DSAs have proved to be the most advanced immune biomarkers in solid organ transplantation in terms of analytical validity, clinical validity and clinical utility. Recent studies are integrating multiple HLA-DSA characteristics including antibody specificity, HLA class, quantity, immunoglobulin G subclass, and complement-binding capacity to improve risk assessment peritransplant, diagnosis and treatment evaluation of antibody-mediated rejection, immune monitoring posttransplant, and transplant prognosis evaluation. In addition, integration of HLA-DSAs to clinical, functional and histological transplant parameters has further consolidated the utility of HLA-DSAs as robust biomarkers and allows to build new tools for monitoring, precision diagnosis, and risk stratification for individual patients. However, prospective and randomized-controlled studies addressing the clinical benefit and cost-effectiveness of HLA-DSA-based monitoring and patient management strategies are required to demonstrate that the use of HLA-DSAs as biomarkers can improve current clinical practice and transplant outcomes.
Collapse
Affiliation(s)
- Kevin Louis
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris
- Human Immunology and Immunopathology, Université de Paris
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| |
Collapse
|
28
|
Naesens M, Budde K, Hilbrands L, Oberbauer R, Bellini MI, Glotz D, Grinyó J, Heemann U, Jochmans I, Pengel L, Reinders M, Schneeberger S, Loupy A. Surrogate Endpoints for Late Kidney Transplantation Failure. Transpl Int 2022; 35:10136. [PMID: 35669974 PMCID: PMC9163814 DOI: 10.3389/ti.2022.10136] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022]
Abstract
In kidney transplant recipients, late graft failure is often multifactorial. In addition, primary endpoints in kidney transplantation studies seek to demonstrate the short-term efficacy and safety of clinical interventions. Although such endpoints might demonstrate short-term improvement in specific aspects of graft function or incidence of rejection, such findings do not automatically translate into meaningful long-term graft survival benefits. Combining many factors into a well-validated model is therefore more likely to predict long-term outcome and better reflect the complexity of late graft failure than using single endpoints. If conditional marketing authorization could be considered for therapies that aim to improve long-term outcomes following kidney transplantation, then the surrogate endpoint for graft failure in clinical trial settings needs clearer definition. This Consensus Report considers the potential benefits and drawbacks of several candidate surrogate endpoints (including estimated glomerular filtration rate, proteinuria, histological lesions, and donor-specific anti-human leukocyte antigen antibodies) and composite scoring systems. The content was created from information prepared by a working group within the European Society for Organ Transplantation (ESOT). The group submitted a Broad Scientific Advice request to the European Medicines Agency (EMA), June 2020: the request focused on clinical trial design and endpoints in kidney transplantation. Following discussion and refinement, the EMA made final recommendations to ESOT in December 2020 regarding the potential to use surrogate endpoints in clinical studies that aim to improving late graft failure.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | | | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, Hôpital Saint Louis, Paris, France
| | | | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Ina Jochmans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Liset Pengel
- Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Marlies Reinders
- Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan Schneeberger
- Department of General, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| |
Collapse
|
29
|
Donovan DJ, Richmond ME, Bacha EA, Addonizio LJ, Zuckerman WA. Association between homograft tissue exposure and allosensitization prior to heart transplant in patients with congenital heart disease. Pediatr Transplant 2022; 26:e14201. [PMID: 34889487 DOI: 10.1111/petr.14201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/01/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Surgical repair for patients with congenital heart disease (CHD) often incorporates homograft tissue or other foreign material that can lead to allosensitization. We sought to identify the relationship between pre-sensitization prior to heart transplant and exposure to homograft tissue in CHD patients. METHODS Retrospective chart review of all CHD patients who underwent heart transplant at a major pediatric transplant center between 1/1/2011-3/31/18. Operative records determined use of homograft tissue or foreign material. Panel reactive antibody (PRA) and LuminexTM single-antigen bead (SAB) testing results were reviewed. Statistical analysis determined odds of pre-sensitization in patients exposed to homograft tissue. RESULTS Fifty-six CHD patients underwent transplant during the review period. Thirteen patients (23%) were pre-sensitized by PRA>10%. By SAB testing, 33 patients (59%) developed any anti-HLA antibody >0 MFI, 30 patients (54%) >2000 MFI, and 19 patients (34%) >6000 MFI. Patients with homografts were more likely to be pre-sensitized by PRA (OR = 7.31, p = .007), and to have developed any anti-HLA antibody at various levels, >0 (OR = 4.52, p = .034), >2000 (OR = 8.59, p = .003), and >6000 (OR = 8.50, p = .004). Of patients with homografts, those pre-sensitized by PRA had longer exposure times (9.80 vs 4.96 years, p = .025). There was no difference in exposure time with relation to pre-sensitization by SAB testing. CONCLUSIONS Previous exposure to homograft tissue appears to increase the odds of pre-sensitization by either the PRA or SAB testing. Longer exposure time to homograft tissue prior to transplant is associated with increased pre-sensitization at transplant as determined by PRA, though not by SAB testing.
Collapse
Affiliation(s)
- Denis J Donovan
- Division of Pediatric Cardiology, New York-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York, USA
| | - Marc E Richmond
- Division of Pediatric Cardiology, New York-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York, USA
| | - Emile A Bacha
- Division of Pediatric Cardiac Surgery, New York-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York, USA
| | - Linda J Addonizio
- Division of Pediatric Cardiology, New York-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York, USA
| | - Warren A Zuckerman
- Division of Pediatric Cardiology, New York-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
30
|
Sigurjonsdottir VK, Purington N, Chaudhuri A, Zhang BM, Fernandez-Vina M, Palsson R, Kambham N, Charu V, Piburn K, Maestretti L, Shah A, Gallo A, Concepcion W, Grimm PC. Complement-Binding Donor-Specific Anti-HLA Antibodies: Biomarker for Immunologic Risk Stratification in Pediatric Kidney Transplantation Recipients. Transpl Int 2022; 35:10158. [PMID: 35992747 PMCID: PMC9386741 DOI: 10.3389/ti.2021.10158] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 01/09/2023]
Abstract
Antibody-mediated rejection is a common cause of early kidney allograft loss but the specifics of antibody measurement, therapies and endpoints have not been universally defined. In this retrospective study, we assessed the performance of risk stratification using systematic donor-specific antibody (DSA) monitoring. Included in the study were children who underwent kidney transplantation between January 1, 2010 and March 1, 2018 at Stanford, with at least 12-months follow-up. A total of 233 patients were included with a mean follow-up time of 45 (range, 9–108) months. Median age at transplant was 12.3 years, 46.8% were female, and 76% had a deceased donor transplant. Fifty-two (22%) formed C1q-binding de novo donor-specific antibodies (C1q-dnDSA). After a standardized augmented immunosuppressive protocol was implemented, C1q-dnDSA disappeared in 31 (58.5%). Graft failure occurred in 16 patients at a median of 54 (range, 5–83) months, of whom 14 formed dnDSA. The 14 patients who lost their graft due to rejection, all had persistent C1q-dnDSA. C1q-binding status improved the individual risk assessment, with persistent; C1q binding yielding the strongest independent association of graft failure (hazard ratio, 45.5; 95% confidence interval, 11.7–177.4). C1q-dnDSA is more useful than standard dnDSA as a noninvasive biomarker for identifying patients at the highest risk of graft failure.
Collapse
Affiliation(s)
- Vaka K. Sigurjonsdottir
- Division of Nephrology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Division of Nephrology, Internal Medicine and Emergency Services, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
- *Correspondence: Vaka K. Sigurjonsdottir,
| | - Natasha Purington
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Palo Alto, CA, United States
| | - Abanti Chaudhuri
- Division of Nephrology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| | - Bing M. Zhang
- Histocompatibility and Immunogenetics Laboratory, Stanford Blood Center, Stanford University, Palo Alto, CA, United States
| | - Marcelo Fernandez-Vina
- Histocompatibility and Immunogenetics Laboratory, Stanford Blood Center, Stanford University, Palo Alto, CA, United States
| | - Runolfur Palsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Division of Nephrology, Internal Medicine and Emergency Services, Landspitali–The National University Hospital of Iceland, Reykjavik, Iceland
| | - Neeraja Kambham
- Department of Pathology, Stanford University, Palo Alto, CA, United States
| | - Vivek Charu
- Department of Pathology, Stanford University, Palo Alto, CA, United States
| | - Kim Piburn
- Division of Nephrology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| | - Lynn Maestretti
- Division of Nephrology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| | - Anika Shah
- Histocompatibility and Immunogenetics Laboratory, Stanford Blood Center, Stanford University, Palo Alto, CA, United States
| | - Amy Gallo
- Division of Nephrology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Palo Alto, CA, United States
| | - Waldo Concepcion
- Transplantation Services, Mohamed Bin Rashid University, Dubai, United Arab Emirates
| | - Paul C. Grimm
- Division of Nephrology, Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
31
|
Mangiola M, Marrari M, Xu Q, Sanchez PG, Zeevi A. Approaching the sensitized lung patient: risk assessment for donor acceptance. J Thorac Dis 2022; 13:6725-6736. [PMID: 34992848 PMCID: PMC8662510 DOI: 10.21037/jtd-2021-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
The presence of HLA antibodies is widely recognized as a barrier to solid organ transplantation, and for lung transplant candidates, it has a significant negative impact on both waiting time and waiting list mortality. Although HLA antibodies have been associated with a broad spectrum of allograft damage, precise characterization of these antibodies in allosensitized candidates may enhance their accessibility to transplant. The introduction of Luminex-based single antigen bead (SAB) assays has significantly improved antibody detection sensitivity and specificity, but SAB alone is not sufficient for risk-stratification. Functional characterization of donor-specific antibodies (DSA) is paramount to increase donor accessibility for allosensitized lung candidates. We describe here our approach to evaluate sensitized lung transplant candidates. By employing state-of-the-art technologies to assess histocompatibility and determine physiological properties of circulating HLA antibodies, we can provide our Clinical Team a better risk assessment for lung transplant candidates and facilitate a "road map" to transplant. The cases presented in this paper illustrate the "individualized steps" taken to determine calculated panel reactive antibodies (cPRA), titer and complement-fixing properties of each HLA antibody present in circulation. When a donor is considered, we can better predict the risk associated with potentially crossing HLA antibodies, thereby allowing the Clinical Team to approach allosensitized lung patients with an individualized medicine approach. To facilitate safe access of sensitized lung transplant candidates to potential donors, a synergy between the histocompatibility laboratory and the Clinical Team is essential. Ultimately, donor acceptance is a decision based on several parameters, leading to a risk-stratification unique for each patient.
Collapse
Affiliation(s)
| | - Marilyn Marrari
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Qingyong Xu
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Pablo G Sanchez
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adriana Zeevi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Ziemann M, Suwelack B, Banas B, Budde K, Einecke G, Hauser I, Heinemann FM, Kauke T, Kelsch R, Koch M, Lachmann N, Reuter S, Seidl C, Sester U, Zecher D. Determination of unacceptable HLA antigen mismatches in kidney transplant recipients. HLA 2021; 100:3-17. [PMID: 34951119 DOI: 10.1111/tan.14521] [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: 11/26/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022]
Abstract
With the introduction of the virtual allocation crossmatch in the Eurotransplant (ET) region in 2023, the determination of unacceptable antigen mismatches (UAM) in kidney transplant recipients is of utmost importance for histocompatibility laboratories and transplant centers. Therefore, a joined working group of members from the German Society for Immunogenetics (Deutsche Gesellschaft für Immungenetik, DGI) and the German Transplantation Society (Deutsche Transplantationsgesellschaft, DTG) revised and updated the previous recommendations from 2015 in light of recently published evidence. Like in the previous version, a wide range of topics is covered from technical issues to clinical risk factors. This review summarizes the evidence about the prognostic value of contemporary methods for HLA antibody detection and identification, as well as the impact of UAM on waiting time, on which these recommendations are based. As no clear criteria could be determined to differentiate potentially harmful from harmless HLA antibodies, the general recommendation is to assign all HLA against which plausible antibodies are found as UAM. There is, however, a need for individualized solutions for highly immunized patients. These revised recommendations provide a list of aspects that need to be considered when assigning UAM to enable a fair and comprehensible procedure and to harmonize risk stratification prior to kidney transplantation between transplant centers. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Malte Ziemann
- Institute of Transfusion Medicine, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Barbara Suwelack
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Klemens Budde
- Medizinische Klinik m. S. Nephrologie, Charité Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Gunilla Einecke
- Clinic for Renal and Hypertensive Disorders, Medizinische Hochschule Hannover, Hannover, Germany
| | - Ingeborg Hauser
- Department of Nephrology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Falko Markus Heinemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Teresa Kauke
- Division of Thoracic Surgery, Hospital of the Ludwig-Maximilians-University München, München, Germany and Transplantation Center, Hospital of the Ludwig-Maximilians-University München, München, Germany
| | - Reinhard Kelsch
- Institute of Transfusion Medicine and Transplantation Immunology, University Hospital Münster, Münster, Germany
| | - Martina Koch
- General-, Visceral- and Transplant Surgery, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Nils Lachmann
- Institute for Transfusion Medicine, H&I Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Reuter
- Medizinische Klinik D, University Hospital Münster, Münster, Germany
| | - Christian Seidl
- Institute for Transfusion Medicine and Immunohaematology, German Red Cross Baden-Württemberg-Hessen, Frankfurt am Main, Germany
| | - Urban Sester
- Transplant center, University Hospital of Saarland, Homburg/Saar, Germany
| | - Daniel Zecher
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
33
|
Charreau B. Cellular and Molecular Crosstalk of Graft Endothelial Cells During AMR: Effector Functions and Mechanisms. Transplantation 2021; 105:e156-e167. [PMID: 33724240 DOI: 10.1097/tp.0000000000003741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Graft endothelial cell (EC) injury is central to the pathogenesis of antibody-mediated rejection (AMR). The ability of donor-specific antibodies (DSA) to bind C1q and activate the classical complement pathway is an efficient predictor of graft rejection highlighting complement-dependent cytotoxicity as a key process operating during AMR. In the past 5 y, clinical studies further established the cellular and molecular signatures of AMR revealing the key contribution of other, IgG-dependent and -independent, effector mechanisms mediated by infiltrating NK cells and macrophages. Beyond binding to alloantigens, DSA IgG can activate NK cells and mediate antibody-dependent cell cytotoxicity through interacting with Fcγ receptors (FcγRs) such as FcγRIIIa (CD16a). FcRn, a nonconventional FcγR that allows IgG recycling, is highly expressed on ECs and may contribute to the long-term persistence of DSA in blood. Activation of NK cells and macrophages results in the production of proinflammatory cytokines such as TNF and IFNγ that induce transient and reversible changes in the EC phenotype and functions promoting coagulation, inflammation, vascular permeability, leukocyte trafficking. MHC class I mismatch between transplant donor and recipient can create a situation of "missing self" allowing NK cells to kill graft ECs. Depending on the microenvironment, cellular proximity with ECs may participate in macrophage polarization toward an M1 proinflammatory or an M2 phenotype favoring inflammation or vascular repair. Monocytes/macrophages participate in the loss of endothelial specificity in the process of endothelial-to-mesenchymal transition involved in renal and cardiac fibrosis and AMR and may differentiate into ECs enabling vessel and graft (re)-endothelialization.
Collapse
Affiliation(s)
- Béatrice Charreau
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et en Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
34
|
Wang M, Patel NJ, Zhang X, Kransdorf EP, Azarbal B, Kittleson MM, Czer LSC, Kobashigawa JA, Patel JK. The effects of donor-specific antibody characteristics on cardiac allograft vasculopathy. Clin Transplant 2021; 35:e14483. [PMID: 34546613 DOI: 10.1111/ctr.14483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) causes late graft dysfunction and post-transplant mortality. Currently, the effects of different donor-specific antibodies (DSA) on the severity of CAV remain unclear. METHOD We evaluated 526 adult heart transplant recipients at a single center between January 2010 and August 2015. Subjects were divided into those with DSA (n = 142) and those without DSA (n = 384, control). The DSA group was stratified into persistent DSA (n = 34), transient DSA (n = 105), 1:8 dilution DSA (n = 45), complement-binding (C1q) DSA (n = 36), Class I DSA (n = 37), and Class II DSA (n = 105). The primary outcome was the incidence of moderate-to-severe CAV (CAV 2/3) at 5-year follow-up. RESULTS Subjects with persistent DSA, 1:8 dilution DSA, and C1q DSA had higher incidence of CAV 2/3 compared the control group (17.6%, 13.3%, and 16.7% vs. 3.1%, respectively; P≤ .001). The incidence of CAV 2/3 between subjects with transient DSA and the control group was similar (2.8% vs. 3.1%; P = .888). Subjects with Class II DSA also had higher incidence of CAV 2/3 (7.6% vs. 3.1%; P = .039). CONCLUSION DSA that are persistent, 1:8 dilution positive, C1q positive, and Class II are associated with more severe grades of CAV. These DSA characteristics may prognosticate disease and warrant consideration for treatment.
Collapse
Affiliation(s)
- Maggie Wang
- Department of Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nikhil J Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Xiaohai Zhang
- HLA and Immunogenetics Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Evan P Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Babak Azarbal
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michelle M Kittleson
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Lawrence S C Czer
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jon A Kobashigawa
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jignesh K Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
35
|
Abstract
Defined as histologic evidence of rejection on a protocol biopsy in the absence of kidney dysfunction, subclinical rejection has garnered attention since the 1990s. The major focus of much of this research, however, has been subclinical T cell-mediated rejection (TCMR). Herein, we review the literature on subclinical antibody-mediated rejection (AMR), which may occur with either preexisting donor-specific antibodies (DSA) or upon the development of de novo DSA (dnDSA). In both situations, subsequent kidney function and graft survival are compromised. Thus, we recommend protocol biopsy routinely within the first year with preexisting DSA and at the initial detection of dnDSA. In those with positive biopsies, baseline immunosuppression should be maximized, any associated TCMR treated, and adherence stressed, but it remains uncertain if antibody-reduction treatment should be initiated. Less invasive testing of blood for donor DNA or gene profiling may have a role in follow-up of those with negative initial biopsies. If a protocol biopsy is positive in the absence of detectable HLA-DSA, it also remains to be determined whether non-HLA-DSA should be screened for either in particular or on a genome-wide basis and how these patients should be treated. Randomized controlled trials are clearly needed.
Collapse
|
36
|
Rossi AP, Alloway RR, Hildeman D, Woodle ES. Plasma cell biology: Foundations for targeted therapeutic development in transplantation. Immunol Rev 2021; 303:168-186. [PMID: 34254320 DOI: 10.1111/imr.13011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022]
Abstract
Solid organ transplantation is a life-saving procedure for patients with end-stage organ disease. Over the past 70 years, tremendous progress has been made in solid organ transplantation, particularly in T-cell-targeted immunosuppression and organ allocation systems. However, humoral alloimmune responses remain a major challenge to progress. Patients with preexisting antibodies to human leukocyte antigen (HLA) are at significant disadvantages in regard to receiving a well-matched organ, moreover, those who develop anti-HLA antibodies after transplantation face a significant foreshortening of renal allograft survival. Historical therapies to desensitize patients prior to transplantation or to treat posttransplant AMR have had limited effectiveness, likely because they do not significantly reduce antibody levels, as plasma cells, the source of antibody production, remain largely unaffected. Herein, we will discuss the significance of plasma cells in transplantation, aspects of their biology as potential therapeutic targets, clinical challenges in developing strategies to target plasma cells in transplantation, and lastly, novel approaches that have potential to advance the field.
Collapse
Affiliation(s)
- Amy P Rossi
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rita R Alloway
- Division of Nephrology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - David Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - E Steve Woodle
- Division of Transplantation, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
37
|
Detection of Antibodies Against Human Leukocyte Antigen Class II in the Sera of Patients Receiving Intravenous Immunoglobulin. Transplant Direct 2021; 7:e697. [PMID: 34036167 PMCID: PMC8133174 DOI: 10.1097/txd.0000000000001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 02/11/2021] [Indexed: 11/25/2022] Open
Abstract
Supplemental Digital Content is available in the text. IVIG is occasionally used for preventing and treating severe infections of patients who are to undergo transplantation. Administration of IVIG, which includes high-titer antibodies (Abs) against HLA class I and II, might have a substantial influence on the HLA Ab test results of these patients. However, this issue has remained unreported.
Collapse
|
38
|
Bertrand D, Kaveri R, Laurent C, Gatault P, Jauréguy M, Garrouste C, Sayegh J, Bouvier N, Caillard S, Lanfranco L, Thierry A, François A, Hau F, Etienne I, Guerrot D, Farce F. Intensity of de novo DSA detected by Immucor Lifecodes assay and C3d fixing antibodies are not predictive of subclinical ABMR after Kidney Transplantation. PLoS One 2021; 16:e0249934. [PMID: 33886604 PMCID: PMC8062066 DOI: 10.1371/journal.pone.0249934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/27/2021] [Indexed: 01/06/2023] Open
Abstract
De novo donor-specific antibodies (dnDSA) are associated with antibody-mediated rejection (ABMR) and allograft loss. We tested Immucor* (IM) Luminex Single-antigen beads (LSAB) assay and C3d-fixing antibodies in the setting of dnDSA and subclinical (s) ABMR. This retrospective multicentric study included 123 patients biopsied because of the presence of subclinical de novo DSA detected by One Lamda* Labscreen (MFI > 1000). In 112 patients, sera of the day of the biopsy were available and tested in a central lab with IM Lifecodes LSAB and C3d fixing antibodies assays. In 16 patients (14.3%), no DSA was detected using Immucor test. In 96 patients, at least one DSA was determined with IM. Systematic biopsies showed active sABMR in 30 patients (31.2%), chronic active sABMR in 17 patients (17.7%) and no lesions of sABMR in 49 KT recipients (51%). Intensitity criteria (BCM, BCR and AD-BCR) of DSA were not statistically different between these 3 histological groups. The proportion of patients with C3d-fixing DSA was not statistically different between the 3 groups and did not offer any prognostic value regarding graft survival. Performing biopsy for dnDSA could not be guided by the intensity criteria of IM LSAB assay. C3d-fixing DSA do not offer added value.
Collapse
Affiliation(s)
- Dominique Bertrand
- Nephrology Kidney Transplantation Dialysis, CHU Rouen, Rouen, France
- * E-mail:
| | | | - Charlotte Laurent
- Nephrology Kidney Transplantation Dialysis, CHU Rouen, Rouen, France
| | | | - Maïté Jauréguy
- Nephrology Kidney Transplantation, CHU Amiens, Amiens, France
| | - Cyril Garrouste
- Nephrology Kidney Transplantation, CHU Clermont Ferrand, Clermont Ferrand, France
| | - Johnny Sayegh
- Nephrology Kidney Transplantation, CHU Angers, Angers, France
| | | | - Sophie Caillard
- Nephrology Kidney Transplantation, CHU Strasbourg, Strasbourg, France
| | - Luca Lanfranco
- Nephrology Kidney Transplantation, CHU Brest, Brest, France
| | - Antoine Thierry
- Nephrology Kidney Transplantation, CHU Poitiers, Poitiers, France
| | | | | | - Isabelle Etienne
- Nephrology Kidney Transplantation Dialysis, CHU Rouen, Rouen, France
| | - Dominique Guerrot
- Nephrology Kidney Transplantation Dialysis, CHU Rouen, Rouen, France
| | | |
Collapse
|
39
|
Bestard O, Couzi L, Crespo M, Kessaris N, Thaunat O. Stratifying the humoral risk of candidates to a solid organ transplantation: a proposal of the ENGAGE working group. Transpl Int 2021; 34:1005-1018. [PMID: 33786891 DOI: 10.1111/tri.13874] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
Detection of circulating antibodies directed against human leukocyte antigen (HLA) molecules, which corresponds to the current definition of 'sensitized patient', has been shown to have a severe impact on both access to transplantation and, if the anti-HLA antibodies are specific to the selected donor, survival of the graft. However, not all donor-specific antibodies (DSA) are equally harmful to the graft and progress in the understanding of humoral memory has led to the conclusion that absence of DSA at transplantation does not rule out the possibility that the patient has a preformed cellular humoral memory against the graft (thereby defining a category of DSA-negative sensitized recipients). Technological progress has led to the generation of new assays that offer unprecedented precision in exploring the different layers (serological and cellular) of alloimmune humoral memory. Based on this recent knowledge, the EuropeaN Guidelines for the mAnagement of Graft rEcipients (ENGAGE) working group to propose an updated definition of sensitization in candidates for solid organ transplantation - one that moves away from the current binary division towards a definition based on homogenous strata with similar humoral risk.
Collapse
Affiliation(s)
- Oriol Bestard
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain.,Bellvitge Research Institute (IDIBELL), Barcelona, Spain
| | - Lionel Couzi
- Nephrology-Transplantation-Dialysis, CHU Bordeaux, Bordeaux, France.,CNRS-UMR 5164 Immuno ConcEpT, Bordeaux University, Bordeaux, France
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar, Barcelona, Spain.,Nephropathies Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Nicos Kessaris
- Department of Nephrology and Transplantation, Guy's Hospital, London, UK.,King's College London, London, UK
| | - Olivier Thaunat
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Lyon, France.,Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| |
Collapse
|
40
|
Koenig A, Mezaache S, Callemeyn J, Barba T, Mathias V, Sicard A, Charreau B, Rabeyrin M, Dijoud F, Picard C, Meas-Yedid V, Olivo-Marin JC, Morelon E, Naesens M, Dubois V, Thaunat O. Missing Self-Induced Activation of NK Cells Combines with Non-Complement-Fixing Donor-Specific Antibodies to Accelerate Kidney Transplant Loss in Chronic Antibody-Mediated Rejection. J Am Soc Nephrol 2021; 32:479-494. [PMID: 33239394 PMCID: PMC8054908 DOI: 10.1681/asn.2020040433] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/06/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Binding of donor-specific antibodies (DSAs) to kidney allograft endothelial cells that does not activate the classic complement cascade can trigger the recruitment of innate immune effectors, including NK cells. Activated NK cells contribute to microvascular inflammation leading to chronic antibody-mediated rejection (AMR). Recipient NK cells can also trigger antibody-independent microvascular inflammation by sensing the absence of self HLA class I molecules ("missing self") on allograft endothelial cells. This translational study investigated whether the condition of missing self amplifies DSA-dependent NK cell activation to worsen chronic AMR. METHODS AND RESULTS Among 1682 kidney transplant recipients who underwent an allograft biopsy at Lyon University Hospital between 2004 and 2017, 135 fulfilled the diagnostic criteria for AMR and were enrolled in the study. Patients with complement-fixing DSAs identified by a positive C3d binding assay (n=73, 54%) had a higher risk of transplant failure (P=0.002). Among the remaining patients with complement-independent chronic AMR (n=62, 46%), those in whom missing self was identified through donor and recipient genotyping exhibited worse allograft survival (P=0.02). In multivariable analysis, only proteinuria (HR: 7.24; P=0.01) and the presence of missing self (HR: 3.57; P=0.04) were independent predictors for transplant failure following diagnosis of chronic AMR. Cocultures of human NK cells and endothelial cells confirmed that addition of missing self to DSA-induced NK cell activation increased endothelial damage. CONCLUSIONS The assessment of missing self at the time of diagnosis of chronic AMR identifies patients at higher risk for kidney transplant failure.
Collapse
Affiliation(s)
- Alice Koenig
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Sarah Mezaache
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Jasper Callemeyn
- Department of Microbiology, Immunology and Transplantation, Catholic University (KU) Leuven, University of Leuven, Leuven, Belgium,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Thomas Barba
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Virginie Mathias
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Human Leukocyte Antigen (HLA) Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Antoine Sicard
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Béatrice Charreau
- Centre de Recherche en Transplantation et Immunologie (CRTI), University Hospital Center (CHU) Nantes, Université de Nantes, National Institute for Health and Medical Research (INSERM), Mixed University Unit (UMR) 1064, Transplantation Urology Nephrology Institute (ITUN), Nantes, France
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | | | - Cécile Picard
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | | | | | - Emmanuel Morelon
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Catholic University (KU) Leuven, University of Leuven, Leuven, Belgium,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Valérie Dubois
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Human Leukocyte Antigen (HLA) Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Olivier Thaunat
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
41
|
Beyzaei Z, Geramizadeh B, Bagheri Z, Karimzadeh S, Shojazadeh A. De Novo Donor Specific Antibody and Long-Term Outcome After Liver Transplantation: A Systematic Review and Meta-Analysis. Front Immunol 2020; 11:613128. [PMID: 33424868 PMCID: PMC7786049 DOI: 10.3389/fimmu.2020.613128] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/13/2020] [Indexed: 12/23/2022] Open
Abstract
Background The impact of de novo anti-HLA donor-specific alloantibodies (DSA) which develop after long-term liver transplantation (LT) remains controversial and unclear. The aim of this study was to investigate the role of de novo DSAs on the outcome in LT. Methods We did a systematic review and meta-analysis of observational studies published until Dec 31, 2019, that reported de novo DSA outcome data (≥1 year of follow-up) after liver transplant. A literature search in the MEDLINE/PubMed, EMBASE, Cochrane Library, Scopus and Web of Science Core Collection databases was performed. Results Of 5,325 studies identified, 15 fulfilled our inclusion criteria. The studies which reported 2016 liver transplant recipients with de novo DSAs showed an increased complication risk, i.e. graft loss and chronic rejection (OR 3.61; 95% CI 1.94-6.71, P < 0.001; I2 58.19%), and allograft rejection alone (OR 6.43; 95% CI: 3.17-13.04; P < 0.001; I2 49.77%); they were compared to patients without de novo DSAs. The association between de novo DSAs and overall outcome failure was consistent across all subgroups and sensitivity analysis. Conclusions Our study suggested that de novo DSAs had a significant deleterious impact on the liver transplant risk of rejection. The routine detection of de novo DSAs may be beneficial as noninvasive biomarker-guided risk stratification.
Collapse
Affiliation(s)
- Zahra Beyzaei
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, Medical School of Shiraz University, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Bagheri
- Department of Biostatistics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Karimzadeh
- Shiraz Medical School Library, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Shojazadeh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
42
|
Oprzędkiewicz A, Mado H, Szczurek W, Gąsior M, Szyguła-Jurkiewicz B. Donor-recipient Matching in Heart Transplantation. Open Cardiovasc Med J 2020. [DOI: 10.2174/18741924020140100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Heart transplantation remains the treatment of choice for end-stage Heart Failure (HF). Due to the shortage of organs for transplantation and the occurrence of perioperative complications, a key problem is donor matching, which should result in increased survival and improved quality of life for patients. The success of this procedure depends on various parameters such as gender, weight, ABO blood group and Human Leukocyte Antigen (HLA) system of both the recipient and the donor. Furthermore, non-HLA antigens may also be valuable in donor-recipient matching. The aim of this article is to summarize the recent knowledge on the impact of various factors on accurate donor-recipient matching to heart transplantation.
Collapse
|
43
|
Hautz T, Messner F, Weissenbacher A, Hackl H, Kumnig M, Ninkovic M, Berchtold V, Krapf J, Zelger BG, Zelger B, Wolfram D, Pierer G, Löscher WN, Zimmermann R, Gabl M, Arora R, Brandacher G, Margreiter R, Öfner D, Schneeberger S. Long-term outcome after hand and forearm transplantation - a retrospective study. Transpl Int 2020; 33:1762-1778. [PMID: 32970891 PMCID: PMC7756600 DOI: 10.1111/tri.13752] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/04/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
Between 2000 and 2014, five patients received bilateral hand (n = 3), bilateral forearm (n = 1), and unilateral hand (n = 1) transplants at the Innsbruck Medical University Hospital. We provide a comprehensive report of the long-term results at 20 years. During the 6-20 years follow-up, 43 rejection episodes were recorded in total. Of these, 27.9% were antibody-related with serum donor-specific alloantibodies (DSA) and skin-infiltrating B-cells. The cell phenotype in rejecting skin biopsies changed and C4d-staining increased with time post-transplantation. In the long-term, a change in hand appearance was observed. The functional outcome was highly depending on the level of amputation. The number and severity of rejections did not correlate with hand function, but negatively impacted on the patients´ well-being and quality of life. Patient satisfaction significantly correlated with upper limb function. One hand allograft eventually developed severe allograft vasculopathy and was amputated at 7 years. The patient later died due to progressive gastric cancer. The other four patients are currently rejection-free with moderate levels of immunosuppression. Hand transplantation remains a therapeutic option for carefully selected patients. A stable immunologic situation with optimized and individually adopted immunosuppression favors good compliance and patient satisfaction and may prevent development of DSA.
Collapse
Affiliation(s)
- Theresa Hautz
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Franka Messner
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Annemarie Weissenbacher
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Hubert Hackl
- Division of BioinformaticsBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Martin Kumnig
- Department of Psychiatry, Psychotherapy and PsychosomaticCenter for Advanced Psychology in Plastic and Transplant SurgeryMedical University of InnsbruckInnsbruckAustria
| | - Marina Ninkovic
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Valeria Berchtold
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Johanna Krapf
- Department of Plastic, Reconstructive and Aesthetic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Bettina G. Zelger
- Department of PathologyMedical University of InnsbruckInnsbruckAustria
| | - Bernhard Zelger
- Department of DermatologyMedical University of InnsbruckInnsbruckAustria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Gerhard Pierer
- Department of Plastic, Reconstructive and Aesthetic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | | | - Robert Zimmermann
- Department of Plastic, Reconstructive and Aesthetic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Markus Gabl
- Department for Trauma SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Rohit Arora
- Department for Trauma SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Gerald Brandacher
- Vascularized Composite Allotransplantation (VCA) LaboratoryDepartment of Plastic and Reconstructive SurgeryJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Raimund Margreiter
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Dietmar Öfner
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic SurgeryCenter of Operative MedicineMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
44
|
Cleary SJ, Kwaan N, Tian JJ, Calabrese DR, Mallavia B, Magnen M, Greenland JR, Urisman A, Singer JP, Hays SR, Kukreja J, Hay AM, Howie HL, Toy P, Lowell CA, Morrell CN, Zimring JC, Looney MR. Complement activation on endothelium initiates antibody-mediated acute lung injury. J Clin Invest 2020; 130:5909-5923. [PMID: 32730229 PMCID: PMC7598054 DOI: 10.1172/jci138136] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Antibodies targeting human leukocyte antigen (HLA)/major histocompatibility complex (MHC) proteins limit successful transplantation and transfusion, and their presence in blood products can cause lethal transfusion-related acute lung injury (TRALI). It is unclear which cell types are bound by these anti-leukocyte antibodies to initiate an immunologic cascade resulting in lung injury. We therefore conditionally removed MHC class I (MHC I) from likely cellular targets in antibody-mediated lung injury. Only the removal of endothelial MHC I reduced lung injury and mortality, related mechanistically to absent endothelial complement fixation and lung platelet retention. Restoration of endothelial MHC I rendered MHC I-deficient mice susceptible to lung injury. Neutrophil responses, including neutrophil extracellular trap (NET) release, were intact in endothelial MHC I-deficient mice, whereas complement depletion reduced both lung injury and NETs. Human pulmonary endothelial cells showed high HLA class I expression, and posttransfusion complement activation was increased in clinical TRALI. These results indicate that the critical source of antigen for anti-leukocyte antibodies is in fact the endothelium, which reframes our understanding of TRALI as a rapid-onset vasculitis. Inhibition of complement activation may have multiple beneficial effects of reducing endothelial injury, platelet retention, and NET release in conditions where antibodies trigger these pathogenic responses.
Collapse
Affiliation(s)
- Simon J. Cleary
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Nicholas Kwaan
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Daniel R. Calabrese
- Department of Medicine, UCSF, San Francisco, California, USA
- Veterans Affairs Healthcare System, San Francisco, California, USA
| | - Beñat Mallavia
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, UCSF, San Francisco, California, USA
| | - John R. Greenland
- Department of Medicine, UCSF, San Francisco, California, USA
- Veterans Affairs Healthcare System, San Francisco, California, USA
| | | | | | - Steven R. Hays
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Ariel M. Hay
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Heather L. Howie
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Pearl Toy
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | | | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark R. Looney
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
45
|
Visentin J, Couzi L, Taupin JL. Clinical relevance of donor-specific antibodies directed at HLA-C: A long road to acceptance. HLA 2020; 97:3-14. [PMID: 33052032 DOI: 10.1111/tan.14106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022]
Abstract
In solid organ transplantation (SOT), the clinical relevance of donor-specific antibodies (DSA) directed at anti-HLA-A, -B, -DR and -DQ antigens is largely recognized while it is still a matter of debate for DSA directed at HLA-C. In this review, we summarize the peculiarities of HLA-C among class I HLA antigens as well as their immunogenicity, which underlie the clinical relevance of HLA-C locus and anti-HLA-C DSA in SOT. Many factors, both intrinsic and extrinsic to the HLA-C gene and HLA-C protein, explain its lower expression in comparison with HLA-A and -B. This lower expression can explain the apparent lower immunogenicity of HLA-C leading to a lower prevalence and strength of anti-HLA-C antibodies. Nevertheless, HLA-C antigens are truly immunogenic and preformed anti-HLA-C DSA are clinically relevant. Indeed, anti-HLA-C DSA are able to bind donor cells and to activate the complement pathway both ex vivo and in vivo. In line with this, numerous clinical studies now show that preformed DSA directed at native HLA-C molecules induce poorer graft outcomes. We then plead for the inclusion of HLA-C in all transplant allocation systems and we propose a strategy to cope with anti-HLA-C DSA in SOT. Beyond SOT, anti-HLA-C antibodies generate a growing interest in the allo-HCT, transfusion and obstetrics fields, while new concepts such as the role of the "missing-self" in solid organ rejection places HLA-C as an inescapable actor in transplant tolerance.
Collapse
Affiliation(s)
- Jonathan Visentin
- CHU de Bordeaux, Laboratoire d'Immunologie et Immunogénétique, Hôpital Pellegrin, Place Amélie Raba Léon, Bordeaux, France.,University of Bordeaux, CNRS, ImmunoConcEpT, Bordeaux, France
| | - Lionel Couzi
- University of Bordeaux, CNRS, ImmunoConcEpT, Bordeaux, France.,CHU de Bordeaux, Service de Néphrologie, Transplantation, Dialyse et Aphérèses, Hôpital Pellegrin, Place Amélie Raba Léon, Bordeaux, France
| | - Jean-Luc Taupin
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, Paris, France.,INSERM, UMR976, and Institut de Recherche saint-Louis, Université de Paris, Paris, France
| |
Collapse
|
46
|
Recommended Treatment for Antibody-mediated Rejection After Kidney Transplantation: The 2019 Expert Consensus From the Transplantion Society Working Group. Transplantation 2020; 104:911-922. [PMID: 31895348 PMCID: PMC7176344 DOI: 10.1097/tp.0000000000003095] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
With the development of modern solid-phase assays to detect anti-HLA antibodies and a more precise histological classification, the diagnosis of antibody-mediated rejection (AMR) has become more common and is a major cause of kidney graft loss. Currently, there are no approved therapies and treatment guidelines are based on low-level evidence. The number of prospective randomized trials for the treatment of AMR is small, and the lack of an accepted common standard for care has been an impediment to the development of new therapies. To help alleviate this, The Transplantation Society convened a meeting of international experts to develop a consensus as to what is appropriate treatment for active and chronic active AMR. The aim was to reach a consensus for standard of care treatment against which new therapies could be evaluated. At the meeting, the underlying biology of AMR, the criteria for diagnosis, the clinical phenotypes, and outcomes were discussed. The evidence for different treatments was reviewed, and a consensus for what is acceptable standard of care for the treatment of active and chronic active AMR was presented. While it was agreed that the aims of treatment are to preserve renal function, reduce histological injury, and reduce the titer of donor-specific antibody, there was no conclusive evidence to support any specific therapy. As a result, the treatment recommendations are largely based on expert opinion. It is acknowledged that properly conducted and powered clinical trials of biologically plausible agents are urgently needed to improve patient outcomes.
Collapse
|
47
|
Tambur AR, Campbell P, Chong AS, Feng S, Ford ML, Gebel H, Gill RG, Kelsoe G, Kosmoliaptsis V, Mannon RB, Mengel M, Reed EF, Valenzuela NM, Wiebe C, Dijke IE, Sullivan HC, Nickerson P. Sensitization in transplantation: Assessment of risk (STAR) 2019 Working Group Meeting Report. Am J Transplant 2020; 20:2652-2668. [PMID: 32342639 PMCID: PMC7586936 DOI: 10.1111/ajt.15937] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/01/2020] [Accepted: 04/20/2020] [Indexed: 01/25/2023]
Abstract
The purpose of the STAR 2019 Working Group was to build on findings from the initial STAR report to further clarify the expectations, limitations, perceptions, and utility of alloimmune assays that are currently in use or in development for risk assessment in the setting of organ transplantation. The goal was to determine the precision and clinical feasibility/utility of such assays in evaluating both memory and primary alloimmune risks. The process included a critical review of biologically driven, state-of-the-art, clinical diagnostics literature by experts in the field and an open public forum in a face-to-face meeting to promote broader engagement of the American Society of Transplantation and American Society of Histocompatibility and Immunogenetics membership. This report summarizes the literature review and the workshop discussions. Specifically, it highlights (1) available assays to evaluate the attributes of HLA antibodies and their utility both as clinical diagnostics and as research tools to evaluate the effector mechanisms driving rejection; (2) potential assays to assess the presence of alloimmune T and B cell memory; and (3) progress in the development of HLA molecular mismatch computational scores as a potential prognostic biomarker for primary alloimmunity and its application in research trial design.
Collapse
Affiliation(s)
- Anat R. Tambur
- Department of SurgeryComprehensive Transplant CenterNorthwestern UniversityChicagoIllinoisUSA
| | - Patricia Campbell
- Department of Laboratory Medicine & PathologyUniversity of AlbertaEdmontonCanada
| | - Anita S. Chong
- Section of TransplantationDepartment of SurgeryThe University of ChicagoChicagoIllinoisUSA
| | - Sandy Feng
- Department of SurgeryUCSF Medical CenterSan FranciscoCaliforniaUSA
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant CenterEmory UniversityAtlantaGeorgiaUSA
| | - Howard Gebel
- Department of PathologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Ronald G. Gill
- Department of ImmunologyUniversity of ColoradoDenverColoradoUSA
| | - Garnett Kelsoe
- Department of ImmunologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | | | - Roslyn B. Mannon
- Department of MedicineDivision of NephrologyUniversity of Alabama School of MedicineBirminghamAlabamaUSA
| | - Michael Mengel
- Department of Laboratory Medicine & PathologyUniversity of AlbertaEdmontonCanada
| | - Elaine F. Reed
- Department of Pathology and Laboratory MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Nicole M. Valenzuela
- Department of Pathology and Laboratory MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chris Wiebe
- Department of MedicineUniversity of ManitobaWinnipegManitobaCanada
| | - I. Esme Dijke
- Department of Laboratory Medicine & PathologyUniversity of AlbertaEdmontonCanada
| | - Harold C. Sullivan
- Department of PathologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Peter Nickerson
- Department of MedicineUniversity of ManitobaWinnipegManitobaCanada
| |
Collapse
|
48
|
Jordan SC, Kucher K, Bagger M, Hockey HU, Wagner K, Ammerman N, Vo A. Intravenous immunoglobulin significantly reduces exposure of concomitantly administered anti-C5 monoclonal antibody tesidolumab. Am J Transplant 2020; 20:2581-2588. [PMID: 32301258 DOI: 10.1111/ajt.15922] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/20/2020] [Accepted: 04/02/2020] [Indexed: 01/25/2023]
Abstract
Awareness of drug-drug interactions is critical in organ transplant recipient management. However, biologic agents interfering with monoclonal antibodies is not widely considered. We report the effect of high-dose intravenous immunoglobulin (IVIg) on safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of the human anti-C5 monoclonal antibody tesidolumab (LFG316) in end-stage renal disease patients awaiting kidney transplant. In this single-center, phase 1, open-label, parallel-group study, 8 patients were assigned to receive either single-dose tesidolumab + IVIg or tesidolumab alone, with 56-day follow-up. Within-group PK parameters were consistent. Mean tesidolumab exposure decreased 34%, clearance increased 63%, and half-life decreased 41% comparing tesidolumab + IVIg to tesidolumab alone. IVIg influence on tesidolumab elimination was most evident in the first 3 weeks. Complete suppression of both total and alternative complement activities was maintained for 4 weeks in the tesidolumab alone group and for 2 weeks in the tesidolumab + IVIg group. Tesidolumab was well tolerated. IVIg infused before tesidolumab affected tesidolumab PK and PD, resulting in a shortened period of full complement activity inhibition. These findings suggest a clinically relevant impact of IVIg on monoclonal antibody clearance and indirectly hint at an IVIg mechanism of action in treating autoimmune diseases and allosensitization by accelerating pathogenic IgG antibody degradation. Trial registration number: NCT02878616.
Collapse
Affiliation(s)
- Stanley C Jordan
- Comprehensive Transplant Center, Transplant Immunotherapy Program Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Klaus Kucher
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Morten Bagger
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Kristina Wagner
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Noriko Ammerman
- Comprehensive Transplant Center, Transplant Immunotherapy Program Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ashley Vo
- Comprehensive Transplant Center, Transplant Immunotherapy Program Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
49
|
Zhao S, Liang T, Zhang C, Shi D, Jiang W, Su C, Hou G. IL-27 Rα + cells promoted allorejection via enhancing STAT1/3/5 phosphorylation. J Cell Mol Med 2020; 24:10756-10767. [PMID: 32761753 PMCID: PMC7521268 DOI: 10.1111/jcmm.15700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 06/23/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022] Open
Abstract
Recently, emerging evidence strongly suggested that the activation of interleukin-27 Receptor α (IL-27Rα) could modulate different inflammatory diseases. However, whether IL-27Rα affects allotransplantation rejection is not fully understood. Here, we investigated the role of IL-27Rα on allorejection both in vivo and in vitro. The skin allotransplantation mice models were established, and the dynamic IL-27Rα/IL-27 expression was detected, and IL-27Rα+ spleen cells adoptive transfer was performed. STAT1/3/5 phosphorylation, proliferation and apoptosis were investigated in mixed lymphocyte reaction (MLR) with recombinant IL-27 (rIL-27) stimulation. Finally, IFN-γ/ IL-10 in graft/serum from model mice was detected. Results showed higher IL-27Rα/IL-27 expression in allografted group compared that syngrafted group on day 10 (top point of allorejection). IL-27Rα+ spleen cells accelerated allograft rejection in vivo. rIL-27 significantly promoted proliferation, inhibited apoptosis and increased STAT1/3/5 phosphorylation of alloreactive splenocytes, and these effects of rIL-27 could be almost totally blocked by JAK/ STAT inhibitor and anti-IL-27 p28 Ab. Finally, higher IL-27Rα+ IFN-γ+ cells and lower IL-27Rα+ IL-10+ cells within allografts, and high IFN-γ/low IL-10 in serum of allorejecting mice were detected. In conclusion, these data suggested that IL-27Rα+ cells apparently promoted allograft rejection through enhancing alloreactive proliferation, inhibiting apoptosis and up-regulating IFN-γ via enhancing STAT pathway. Blocking IL-27 pathway may favour to prevent allorejection, and IL-27Rα may be as a high selective molecule for targeting diagnosis and therapy for allotransplantation rejection.
Collapse
Affiliation(s)
- Shanshan Zhao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ting Liang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chao Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dai Shi
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wen Jiang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Su
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guihua Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
50
|
Early conversion to a CNI-free immunosuppression with SRL after renal transplantation-Long-term follow-up of a multicenter trial. PLoS One 2020; 15:e0234396. [PMID: 32756556 PMCID: PMC7406080 DOI: 10.1371/journal.pone.0234396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction Early conversion to a CNI-free immunosuppression with SRL was associated with an improved 1- and 3- yr renal function as compared with a CsA-based regimen in the SMART-Trial. Mixed results were reported on the occurrence of donor specific antibodies under mTOR-Is. Here, we present long-term results of the SMART-Trial. Methods and materials N = 71 from 6 centers (n = 38 SRL and n = 33 CsA) of the original SMART-Trial (ITT n = 140) were enrolled in this observational, non-interventional extension study to collect retrospectively and prospectively follow-up data for the interval since baseline. Primary objective was the development of dnDSA. Blood samples were collected on average 8.7 years after transplantation. Results Development of dnDSA was not different (SRL 5/38, 13.2% vs. CsA 9/33, 27.3%; P = 0.097). GFR remained improved under SRL with 64.37 ml/min/1.73m2 vs. 53.19 ml/min/1.73m2 (p = 0.044). Patient survival did not differ between groups at 10 years. There was a trend towards a reduced graft failure rate (11.6% SRL vs. 23.9% CsA, p = 0.064) and less tumors under SRL (2.6% SRL vs. 15.2% CsA, p = 0.09). Conclusions An early conversion to SRL did not result in an increased incidence of dnDSA nor increased long-term risk for the recipient. Transplant function remains improved with benefits for the graft survival.
Collapse
|