1
|
Yamazaki T, Iwasaki K, Tomono S, Imai M, Miwa Y, Shizuku M, Ashimine S, Ishiyama K, Inui M, Okuzaki D, Okada M, Kobayashi T, Akashi-Takamura S. Human RP105 monoclonal antibody enhances antigen-specific antibody production in unique culture conditions. iScience 2024; 27:110649. [PMID: 39246445 PMCID: PMC11380396 DOI: 10.1016/j.isci.2024.110649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/16/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024] Open
Abstract
Detecting antibodies, particularly those targeting donor human leukocyte antigens in organ transplantation and self-antigens in autoimmune diseases, is crucial for diagnosis and therapy. Radioprotective 105 (RP105), a Toll-like receptor family protein, is expressed in immune-competent cells, such as B cells. Studies in mice have shown that the anti-mouse RP105 antibody strongly activates B cells and triggers an adjuvant effect against viral infections. However, the anti-human RP105 antibody (ɑhRP105) weakly activates human B cells. This study established new culture conditions under, which human B cells are strongly activated by the ɑhRP105. When combined with CpGDNA, specific antibody production against blood group carbohydrates, ɑGal, and SARS-CoV-2 was successfully detected in human B cell cultures. Furthermore, comprehensive analysis using liquid chromatography-electrospray ionization tandem mass spectrometry, single-cell RNA sequencing, and quantitative real-time PCR revealed that ɑhRP105 triggered a different activation stimulus compared to CpGDNA. These findings could help identify antibody-producing B cells in cases of transplant rejection and autoimmune diseases.
Collapse
Affiliation(s)
- Tatsuya Yamazaki
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Kenta Iwasaki
- Department of Kidney Diseases and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Susumu Tomono
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Masaki Imai
- Department of Medical Technology and Sciences, Kyoto Tachibana University, Kyoto, Kyoto, Japan
| | - Yuko Miwa
- Department of Kidney Diseases and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Masato Shizuku
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Satoshi Ashimine
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Kohei Ishiyama
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Masanori Inui
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI-IFReC, Osaka University, Suita, Osaka, Japan
| | - Manabu Okada
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Aichi, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Sachiko Akashi-Takamura
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| |
Collapse
|
2
|
Jiang S, Su H. Exploration of the shared gene signatures and biological mechanisms between ischemia-reperfusion injury and antibody-mediated rejection in renal transplantation. Transpl Immunol 2024; 83:102001. [PMID: 38266883 DOI: 10.1016/j.trim.2024.102001] [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: 06/17/2023] [Revised: 12/22/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Antibody-mediated rejection (ABMR) plays a crucial role in graft loss during allogeneic renal transplantation. In renal transplantation, ischemia-reperfusion injury (IRI) is unavoidable, serves as a major contributor to acute rejection, and is linked to graft loss. However, the mechanisms underlying IRI and ABMR are unclear. Therefore, this study aimed to investigate the shared genetic characteristics and biological mechanisms between IRI and ABMR. METHODS Gene expressions for IRI (GSE43974) and ABMR (GSE129166 and GSE36059) were retrieved from the Gene Expression Omnibus database. The shared differentially expressed genes (DEGs) of IRI and ABMR were identified, and subsequent functional enrichment analysis was performed. Immune cell infiltration in ABMR and its relationship with the shared DEGs were investigated using the CIBERSORT method. Random forest analysis, a protein-protein interaction network, and Cytoscape were used to screen hub genes, which were subsequently subjected to gene set enrichment analysis, miRNA prediction, and transcription factors analysis. The survival analysis was performed through Kaplan-Meier curves. Finally, drug compound prediction was performed on the shared DEGs using the Drug Signature Database. RESULTS Overall, 27 shared DEGs were identified between the renal IRI and ABMR groups. Among these, 24 genes exhibited increased co-expression, whereas none showed decreased co-expression. The shared DEGs were primarily enriched in the inflammation signaling pathways. Notably, CD4 memory T cells were identified as potential critical mediators of IRI, leading to ABMR. Tumor necrosis factor alpha-induced protein 3 (TNFAIP3), interferon regulatory factor 1 (IRF1), and early growth response 2 (EGR2) were identified as key components in the potential mechanism that link IRI and ABMR. Patients undergoing renal transplantation with higher expression levels of TNFAIP3, IRF1, and EGR2 exhibited decreased survival rates compared to those with lower expression levels. CONCLUSION Inflammation is a key mechanism that links IRI and ABMR, with a potential role played by CD4 memory T cells. Furthermore, TNFAIP3, IRF1, and EGR2 are implicated in the underlying mechanism between IRI and ABMR.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
3
|
Valentin MO, Crespo M, Fernandez C, Muro M, Vega R, Palou E, Ruiz JC, Diekman F, Padilla M, Mancebo E, Perez I, Andres A, Ontañon J, Dominguez-Gil B. Improving the Access of Highly Sensitized Patients to Kidney Transplantation From Deceased Donors: The Spanish PATHI Program With Allocation Based on the Virtual Crossmatch. Transplantation 2024; 108:787-801. [PMID: 37867239 DOI: 10.1097/tp.0000000000004824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
BACKGROUND In 2015, the Spanish National Transplant Organization developed a prioritization system (Program for Access to Transplantation for Highly Sensitized Patients [PATHI]) to increase transplant options for patients with calculated panel-reactive antibodies (cPRAs) ≥98%, based on virtual crossmatch. We describe the experience with the implementation of PATHI and assess its efficacy. METHODS PATHI registry was used to collect characteristics of donors and patients between June 15, 2015, and March 1, 2018. One-year graft and patient survival and acute rejection were also measured. A Cox model was used to identify factors related to patient death and graft loss and logistical regression for those associated with rejection. RESULTS One thousand eighty-nine patients were included, and 272 (25%) were transplanted. Transplant rate by cPRA was 54.9%, 40.5%, and 12.8% in patients with cPRA98%, cPRA99%, and cPRA100%, respectively. One-year patient survival was 92.5%. Recipient age ≥60, time under dialysis >7 y, and delayed graft function were mortality risk factors. One-year graft survival was 88.7%. The factor related to graft loss was delayed graft function. The rejection rate was 22%. Factors related to rejection were sex, older recipients, and posttransplant donor-specific antibodies. CONCLUSIONS A prioritization approach increases transplant options for highly sensitized patients with appropriate short-term postransplant outcomes. Along with other programs, PATHI may inspire other countries to adopt strategies to meet transplant needs of these patients.
Collapse
Affiliation(s)
- Maria O Valentin
- Nephrology Department, Hospital Universitario Marqués de Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Marta Crespo
- Nephrology Department, Hospital De Mar, Barcelona, Spain
| | - Constantino Fernandez
- Nephrology Department, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Manuel Muro
- Immunology Department, Hospital Clinico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Rocio Vega
- Transplant Coordination Department, Organización Nacional de Trasplantes, Madrid, Spain
| | - Eduard Palou
- Immunology Department, Hospital Clinic, Barcelona, Spain
| | - Juan Carlos Ruiz
- Nephrology Department, Hospital Universitario Marqués de Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Fritz Diekman
- Nephrology Department, Hospital Clinic, Barcelona, Spain
| | - Maria Padilla
- Immunology Department, Hospital Clinico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Esther Mancebo
- Immunology Department, Hospital 12 de Octubre, Madrid, Spain
| | - Isabel Perez
- Nephrology Department, Hospital Clinico San Carlos, Madrid, Spain
| | - Amado Andres
- Nephrology Department, Hospital 12 de Octubre, Madrid, Spain
| | - Jesus Ontañon
- Immunology Department, Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Beatriz Dominguez-Gil
- Immunology Department, Hospital Clinico Universitario Virgen de la Arrixaca, Murcia, Spain
| |
Collapse
|
4
|
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
|
5
|
Lin J, Lv J, Yu X, Xue X, Yu S, Wang H, Chen J. Single-Cell Heterogeneity Restorative Chimeric Engineering Nanoparticles for Alleviating Antibody-Mediated Allograft Injury. ACS APPLIED MATERIALS & INTERFACES 2023; 15:34588-34606. [PMID: 37459593 DOI: 10.1021/acsami.3c06885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Disturbance of single-cell transcriptional heterogeneity is an inevitable consequence of persistent donor-specific antibody (DSA) production and allosensitization. However, identifying and efficiently clearing allospecific antibody repertoires to restore single-cell transcriptional profiles remain challenging. Here, inspired by the high affinity of natural bacterial proteins for antibodies, a genetic engineered membrane-coated nanoparticle termed as DSA trapper by the engineering chimeric gene of protein A/G with phosphatidylserine ligands for macrophage phagocytosis was reported. It has been shown that DSA trappers adsorbed alloreactive antibodies with high saturation and activated the heterophagic clearance of antibody complexes, alleviating IgG deposition and complement activation. Remarkably, DSA trappers increased the endothelial protective lineages by 8.39-fold, reversed the highly biased cytotoxicity, and promoted the proliferative profiles of Treg cells, directly providing an obligate immune tolerant niche for single-cell heterogeneity restoration. In the mice of allogeneic transplantation, the DSA trapper spared endothelial from inflammatory degenerative rosette, improved the glomerular filtration rate, and prolonged the survival of allogeneic mice from 23.6 to 78.3 days. In general, by identifying the lineage characteristics of rejection-related antibodies, the chimeric engineered DSA trapper realized immunoadsorption and further phagocytosis of alloantibody complexes to restore the single-cell genetic architecture of the allograft, offering a promising prospect for the treatment of alloantibody-mediated immune injury.
Collapse
Affiliation(s)
- Jinwen Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases. Institute of Nephrology, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Zhejiang University, Hangzhou 310003, Zhejiang Province, P. R. China
| | - Junhao Lv
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases. Institute of Nephrology, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Zhejiang University, Hangzhou 310003, Zhejiang Province, P. R. China
| | - Xianping Yu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases. Institute of Nephrology, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Zhejiang University, Hangzhou 310003, Zhejiang Province, P. R. China
| | - Xing Xue
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, P. R. China
| | - Shiping Yu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases. Institute of Nephrology, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Zhejiang University, Hangzhou 310003, Zhejiang Province, P. R. China
| | - Huiping Wang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases. Institute of Nephrology, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Zhejiang University, Hangzhou 310003, Zhejiang Province, P. R. China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases. Institute of Nephrology, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Zhejiang University, Hangzhou 310003, Zhejiang Province, P. R. China
| |
Collapse
|
6
|
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
|
7
|
Bertazza Partigiani N, Negrisolo S, Carraro A, Marzenta D, Manaresi E, Gallinella G, Barzon L, Benetti E. Pre-Existing Intrarenal Parvovirus B19 Infection May Relate to Antibody-Mediated Rejection in Pediatric Kidney Transplant Patients. Int J Mol Sci 2023; 24:ijms24119147. [PMID: 37298109 DOI: 10.3390/ijms24119147] [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: 04/03/2023] [Revised: 05/01/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Viral infections can lead to transplant dysfunction, and their possible role in rejection is described. In total, 218 protocol biopsies performed in 106 children at 6, 12 and 24 months after transplantation were analyzed according to Banff '15. RT-PCR for cytomegalovirus, Epstein-Barr virus, BK virus and Parvovirus B19 was performed on blood and bioptic samples at the time of transplant and each protocol biopsy. The prevalence of intrarenal viral infection increases between 6 and 12 months after transplantation (24% vs. 44%, p = 0.007). Intrarenal Parvovirus B19 infection is also associated with antibody-mediated rejection (ABMR) (50% ABMR vs. 19% T-cell-mediated rejection, p = 0.04). Moreover, Parvovirus infection is higher at 12 months of follow-up and it decreases at 48 months (40.4% vs. 14%, p = 0.02), while in 24% of grafts, Parvovirus is already detectable at the moment of transplantation. Intrarenal Parvovirus B19 infection seems to be related to ABMR in pediatric kidney recipients. The graft itself may be the way of transmission for Parvovirus, so performance of a PCR test for Parvovirus B19 should be considered to identify high-risk patients. Intrarenal Parvovirus infection presents mainly during the first-year post-transplantation; thus, we recommend an active surveillance of donor-specific antibodies (DSA) in patients with intrarenal Parvovirus B19 infection during this period. Indeed, it should be considered a treatment with intravenous immunoglobulins in patients with intrarenal Parvovirus B19 infection and DSA positivity, even in the absence of ABMR criteria for kidney biopsy.
Collapse
Affiliation(s)
- Nicola Bertazza Partigiani
- Pediatric Nephrology, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
- Department of Women's and Children's Health, University of Padua, 35128 Padua, Italy
| | - Susanna Negrisolo
- Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, University of Padova, 35127 Padua, Italy
- Pediatric Research Institute "IRP Città della Speranza", 35127 Padua, Italy
| | - Andrea Carraro
- Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, University of Padova, 35127 Padua, Italy
| | - Diana Marzenta
- Pediatric Nephrology, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
- Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, University of Padova, 35127 Padua, Italy
| | - Elisabetta Manaresi
- Department of Pharmacy and Biotechnology, University of Bologna, 40138 Bologna, Italy
| | - Giorgio Gallinella
- Department of Pharmacy and Biotechnology, University of Bologna, 40138 Bologna, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Elisa Benetti
- Pediatric Nephrology, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy
- Laboratory of Immunopathology and Molecular Biology of the Kidney, Department of Women's and Children's Health, University of Padova, 35127 Padua, Italy
- Pediatric Research Institute "IRP Città della Speranza", 35127 Padua, Italy
| |
Collapse
|
8
|
Kim HW, Lee J, Heo SJ, Kim BS, Huh KH, Yang J. Comparison of high-dose IVIG and rituximab versus rituximab as a preemptive therapy for de novo donor-specific antibodies in kidney transplant patients. Sci Rep 2023; 13:7682. [PMID: 37169835 PMCID: PMC10175554 DOI: 10.1038/s41598-023-34804-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023] Open
Abstract
De novo donor-specific antibody (dnDSA) is associated with a higher risk of kidney graft failure. However, it is unknown whether preemptive treatment of subclinical dnDSA is beneficial. Here, we assessed the efficacy of high-dose intravenous immunoglobulin (IVIG) and rituximab combination therapy for subclinical dnDSA. An open-label randomized controlled clinical trial was conducted at two Korean institutions. Adult (aged ≥ 19 years) kidney transplant patients with subclinical class II dnDSA (mean fluorescence intensity ≥ 1000) were enrolled. Eligible participants were randomly assigned to receive rituximab or rituximab with IVIG at a 1:1 ratio. The primary endpoint was the change in dnDSA titer at 3 and 12 months after treatment. A total of 46 patients (24 for rituximab and 22 for rituximab with IVIG) were included in the analysis. The mean baseline estimated glomerular filtration rate was 66.7 ± 16.3 mL/min/1.73 m2. The titer decline of immune-dominant dnDSA at 12 months in both the preemptive groups was significant. However, there was no difference between the two groups at 12 months. Either kidney allograft function or proteinuria did not differ between the two groups. No antibody-mediated rejection occurred in either group. Preemptive treatment with high-dose IVIG combined with rituximab did not show a better dnDSA reduction compared with rituximab alone.Trial registration: IVIG/Rituximab versus Rituximab in Kidney Transplant With de Novo Donor-specific Antibodies (ClinicalTrials.gov Identifier: NCT04033276, first trial registration (26/07/2019).
Collapse
Affiliation(s)
- Hyung Woo Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Juhan Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seok-Jae Heo
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
| | - Beom Seok Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Kyu Ha Huh
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea.
| | - Jaeseok Yang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Gniewkiewicz M, Czerwinska K, Zielniok K, Durlik M. Impact of Resolved Preformed, Persistent Preformed, and De Novo Anti-HLA Donor-Specific Antibodies in Kidney Transplant Recipients on Long-Term Renal Graft Outcomes. J Clin Med 2023; 12:jcm12103361. [PMID: 37240467 DOI: 10.3390/jcm12103361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The post-transplant evolution of antihuman leukocyte antigen donor-specific antibodies (anti-HLA DSAs) includes three clinical patterns: resolved preformed DSAs, persistent preformed DSAs, and de novo DSAs. The aim of this retrospective study was to analyze the impact of resolved preformed, persistent preformed, and de novo anti-HLA-A, -B, and -DR DSAs in kidney transplant recipients on long-term renal allograft outcomes. This is a post hoc analysis of the study conducted in our transplant center. One hundred eight kidney transplant recipients were included in the study. Patients were followed for a minimum of 24 months after allograft biopsy, which was performed 3 to 24 months after kidney transplantation. The identification of persistent preformed DSAs at the time of biopsy was the most significant predictor of the combined endpoint of the study (>30% decline in estimated glomerular filtration rate or death-censored graft loss; HR = 5.96, 95% CI 2.041-17.431, p = 0.0011), followed by the occurrence of de novo DSAs (HR = 4.48, 95% CI 1.483-13.520, p = 0.0079). No increased risk was observed in patients with resolved preformed DSAs (HR = 1.10, 95% CI 0.139-8.676, p = 0.9305). Patients with resolved preformed DSAs have similar graft prognoses as patients without DSAs, therefore, the persistence of preformed DSAs and development of de novo DSAs are associated with inferior long-term allograft outcomes.
Collapse
Affiliation(s)
- Michal Gniewkiewicz
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| | - Katarzyna Czerwinska
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| | - Katarzyna Zielniok
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| | - Magdalena Durlik
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| |
Collapse
|
10
|
Ono K, Ide K, Nakano R, Sakai H, Tanimine N, Tahara H, Ohira M, Tanaka Y, Ohdan H. Genetic Polymorphisms in Follicular Helper T Cell-Related Molecules Predispose Patients to De Novo Donor-Specific Antibody Formation After Kidney Transplantation. Transplant Proc 2023:S0041-1345(23)00143-4. [PMID: 37031038 DOI: 10.1016/j.transproceed.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Risk prediction of de novo donor-specific antibody (dnDSA) formation is crucial for understanding the long-term prognostic impact of kidney transplantation (KT). Recently, follicular helper T (Tfh) cells, a subtype of CD4+ T cells, have been reported to play an important role in dnDSA formation after solid organ transplantation. Given the growing recognition of the importance of Tfh cells in generating a strong humoral immune response, we examined whether polymorphisms in Tfh cell-related molecules were associated with dnDSA formation after KT. METHODS Eighty-three patients who underwent living-donor KT between January 2013 and February 2020 at Hiroshima University Hospital were included in the study. Six Tfh cell-related molecules (BCL6, CXCR5, CXCL13, ICOS, CD40L, and IL21) that are important for Tfh cell differentiation and maturation in secondary lymphoid tissues were investigated. CTLA4, which is important for Tfh-cell activation, was also investigated. Single nucleotide polymorphisms (SNPs) in the genes for these molecules were detected using Taq Man SNP genotyping and evaluated for their association with dnDSA formation after KT. RESULTS Of the 83 KT recipients, 8 developed dnDSAs during the observation period. No statistically significant differences were observed in the baseline characteristics between patients with and without dnDSA formation, except for donor age. Among the 7 Tfh cell-related molecules, the incidence of dnDSA formation was associated with CXCR5 and CTLA4 SNPs. Furthermore, combining these 2 SNPs enabled more significant stratification of dnDSA formation. CONCLUSION Our findings indicate that genetic polymorphisms in Tfh cell-related molecules are predisposing factors for dnDSA formation after KT.
Collapse
Affiliation(s)
- Kosuke Ono
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kentaro Ide
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Ryosuke Nakano
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Sakai
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Naoki Tanimine
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Tahara
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masahiro Ohira
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuka Tanaka
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastrointestinal and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
11
|
Danger R, Le Berre L, Cadoux M, Kerleau C, Papuchon E, Mai HL, Nguyen TVH, Guérif P, Morelon E, Thaunat O, Legendre C, Anglicheau D, Lefaucheur C, Couzi L, Del Bello A, Kamar N, Le Quintrec M, Goutaudier V, Renaudin K, Giral M, Brouard S. Subclinical rejection-free diagnostic after kidney transplantation using blood gene expression. Kidney Int 2023; 103:1167-1179. [PMID: 36990211 DOI: 10.1016/j.kint.2023.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/16/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
We previously established a six-gene-based blood score associated with operational tolerance in kidney transplantation which was decreased in patients developing anti-HLA donor-specific antibodies (DSA). Herein, we aimed to confirm that this score is associated with immunological events and risk of rejection. We measured this using quantitative PCR (qPCR) and NanoString methods from an independent multicenter cohort of 588 kidney transplant recipients with paired blood samples and biopsies at one year after transplantation validating its association with pre-existing and de novo DSA. From 441 patients with protocol biopsy, there was a significant decrease of the score of tolerance in 45 patients with biopsy-proven subclinical rejection (SCR), a major threat associated with pejorative allograft outcomes that prompted an SCR score refinement. This refinement used only two genes, AKR1C3 and TCL1A, and four clinical parameters (previous experience of rejection, previous transplantation, sex of recipient and tacrolimus uptake). This refined SCR score was able to identify patients unlikely to develop SCR with a C-statistic of 0.864 and a negative predictive value of 98.3%. The SCR score was validated in an external laboratory, with two methods (qPCR and NanoString), and on 447 patients from an independent and multicenter cohort. Moreover, this score allowed reclassifying patients with discrepancies between the DSA presence and the histological diagnosis of antibody mediated rejection unlike kidney function. Thus, our refined SCR score could improve detection of SCR for closer and noninvasive monitoring, allowing early treatment of SCR lesions notably for patients DSA-positive and during lowering of immunosuppressive treatment.
Collapse
Affiliation(s)
- Richard Danger
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France.
| | - Ludmilla Le Berre
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Marion Cadoux
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Clarisse Kerleau
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Emmanuelle Papuchon
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France; Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), CHU Nantes, Nantes, France
| | - Hoa Le Mai
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Thi-Van-Ha Nguyen
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Pierrick Guérif
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Emmanuel Morelon
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, INSERM Unit 1111, Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, INSERM Unit 1111, Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Christophe Legendre
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Necker-Enfants Malades Institute, INSERM, Paris University, Paris, France
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Necker-Enfants Malades Institute, INSERM, Paris University, Paris, France
| | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, INSERM UMR S970, Université Paris Cité, Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis, and Apheresis, CHU Bordeaux, Bordeaux, France
| | - Arnaud Del Bello
- Department of Nephrology and Organ Transplantation, Centre Hospitalier Universitaire de Toulouse, INSERM UMR1291 - Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, Centre Hospitalier Universitaire de Toulouse, INSERM UMR1291 - Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Moglie Le Quintrec
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospital of Lapeyronie, Montpellier, France
| | - Valentin Goutaudier
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Necker-Enfants Malades Institute, INSERM, Paris University, Paris, France; Université Paris Cité, INSERM U970, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Karine Renaudin
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France; CHU Nantes, Service d'Anatomie et Cytologie Pathologiques, Nantes, France
| | - Magali Giral
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France; Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), CHU Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes Université, Nantes, France
| | - Sophie Brouard
- CHU Nantes, Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France; Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), CHU Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes Université, Nantes, France.
| |
Collapse
|
12
|
Gniewkiewicz M, Czerwinska K, Zielniok K, Durlik M. Association of Circulating Anti-HLA Donor-Specific Antibodies and Their Characteristics, including C1q-Binding Capacity, in Kidney Transplant Recipients with Long-Term Renal Graft Outcomes. J Clin Med 2023; 12:jcm12041312. [PMID: 36835848 PMCID: PMC9962721 DOI: 10.3390/jcm12041312] [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: 01/03/2023] [Revised: 02/02/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023] Open
Abstract
Post-transplant antihuman leukocyte antigen donor-specific antibodies (anti-HLA DSAs) monitoring in kidney transplant recipients remains unclear and is currently under investigation. The pathogenicity of anti-HLA DSAs is determined by antibody classes, specificity, mean fluorescent intensity (MFI), C1q-binding capacity, and IgG subclasses. The aim of this study was to investigate the association of circulating DSAs and their characteristics with renal allograft long-term outcomes. The study included 108 consecutive patients from our transplant center who underwent kidney allograft biopsy between November 2018 and November 2020, 3 to 24 months after kidney transplantation. At the time of biopsy, patients' sera were collected for analysis of anti-HLA DSAs. Patients were followed for a median time of 39.0 months (Q1-Q3, 29.8-45.0). Detection of anti-HLA DSAs at the time of biopsy (HR = 5.133, 95% CI 2.150-12.253, p = 0.0002) and their C1q-binding capacity (HR = 14.639, 95% CI 5.320-40.283, p ≤ 0.0001) were independent predictors of the composite of sustained 30% reduction from estimated glomerular filtration rate or death-censored graft failure. Identification of anti-HLA DSAs and their C1q-binding capacity could be useful in identifying kidney transplant recipients at risk for inferior renal allograft function and graft failure. Analysis of C1q is noninvasive, accessible, and should be considered in clinical practice in post-transplant monitoring.
Collapse
Affiliation(s)
- Michal Gniewkiewicz
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
- Correspondence:
| | - Katarzyna Czerwinska
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| | - Katarzyna Zielniok
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| | - Magdalena Durlik
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
| |
Collapse
|
13
|
Zhou S, Ma X, Ma C, Zhu Q, Zhao Q, Li L, Li S, Tang Z, He J, Wu D, Wu X, Qin S. Splenic irradiation combined with plasmapheresis and rituximab: a new option reducing donor-specific antibody in haploidentical hematopoietic stem cell transplantation. Bone Marrow Transplant 2023; 58:226-228. [PMID: 36402923 DOI: 10.1038/s41409-022-01868-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022]
Affiliation(s)
- Shiyuan Zhou
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, Jiangsu Province, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiao Ma
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, Jiangsu Province, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chao Ma
- Soochow Hopes Hematonosis Hospital, Suzhou, Jiangsu Province, China
| | - Qian Zhu
- Soochow Hopes Hematonosis Hospital, Suzhou, Jiangsu Province, China
| | - Qi Zhao
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Ling Li
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shijia Li
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zaixiang Tang
- School of Public Health, Medical College of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Jun He
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, Jiangsu Province, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Depei Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China. .,National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, Jiangsu Province, China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Xiaojin Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China. .,National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, Jiangsu Province, China. .,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Songbing Qin
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
14
|
Daloul R, Sureshkumar K, Schnelle K, Von Stein L, Logan A, Pesavento T. Kidney transplant from HCV viremic donors to HCV-negative recipients and risk for de novo donor specific antibodies and acute rejection. Clin Transplant 2023; 37:e14895. [PMID: 36580971 DOI: 10.1111/ctr.14895] [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: 06/30/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Kidney transplantation from HCV-viremic donors into uninfected recipients is associated with excellent short-term outcomes. However, concerns regarding an increased risk for the development of de novo donor specific antibodies (DSA) and acute rejection have been raised in single center reports. METHODS A retrospective study of HCV-negative kidney-only transplant recipients between 2018 and 2020. Patients were grouped based on the donor HCV status into group 1; HCV-viremic donors, and group 2; HCV-negative donors. Inverse probability of treatment weighting (IPTW), with weights derived from the propensity score, were used to estimate the effect of donors' HCV-viremia on the recipients. The primary objective was to compare the 1-year incidence of de novo DSA. Secondary outcomes included group comparison of the incidence of biopsy proven acute rejection (BPAR), 1-year patient and allograft survival, and 1-year renal allograft function. RESULTS A total of 71 patients were included in the HCV NAT+ group, and 440 in the HCV- negative group. One-year incidence of de novo DSA was higher in the HCV NAT+ group in the IPTW weighted analysis (19% vs. 9%, p = .02). In the unweighted analysis, BPAR occurred in 7% of recipients in the HCV NAT+ group, compared to 3% in the control group (p = .06). However, due to the low event rate in the in the IPTW weighted groups, a statistical significance test could not be performed. Average estimated GFR was higher in the HCV-viremic group at 3 months (61 vs. 53 ml/min/1.73 m2 p = .002), but comparable at 6 (59 vs. 56 ml/min/1.73 m2 , p = .31) and 12 months (60 vs. 55 ml/min/1.73 m2 , p = .07). Patient and allograft survival were comparable between the two groups. CONCLUSION Kidney transplant from HCV-viremic donors was associated with an increased risk for the development of post-transplant de novo DSA in the first year after transplantation, but no difference in patient and graft survival.
Collapse
Affiliation(s)
- Reem Daloul
- Division of Nephrology, Department of Internal Medicine, Allegheny General Hospital, Drexel University College of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kalathil Sureshkumar
- Division of Nephrology, Department of Internal Medicine, Allegheny General Hospital, Drexel University College of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kendra Schnelle
- Department of Pharmacy, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Lauren Von Stein
- Department of Pharmacy, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - April Logan
- Division of Transplantation, Department of Surgery, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Todd Pesavento
- Division of Nephrology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
15
|
Grimaldi V, Pagano M, Moccia G, Maiello C, De Rosa P, Napoli C. Novel insights in the clinical management of hyperimmune patients before and after transplantation. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100056. [PMID: 36714552 PMCID: PMC9876744 DOI: 10.1016/j.crimmu.2023.100056] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Despite improvements in anti-Human Leucocyte Antigens antibody detection, identification, and characterization offer a better in peri-operative management techniques, antibodies remain a serious cause of morbidity and mortality for patients both before and after organ transplantation. Hyperimmune patients are disadvantaged by having to wait longer to receive an organ from a suitably matched donor. They could benefit from desensitization protocols in both pre- and post-transplantation period. Clinical studies are underway to highlight which best desensitization strategies could be assure the best outcome in both heart and kidney transplantation. Although most clinical evidence about desensitization strategies by using anti-CD20 monoclonal antibodies, proteasome inhibitors, anti-CD38 monoclonal antibodies, interleukin-6 blockade, cysteine protease and complement inhibitors, comes from kidney transplantation studies, many of the debated novel concepts can be easily applied to desensitization also in heart transplantation. Here, we discuss the candidates and recipients' management by using most common standard of care and novel therapeutics, desensitization endpoints, and strategies for future studies.
Collapse
Affiliation(s)
- Vincenzo Grimaldi
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy,Corresponding author.
| | - Martina Pagano
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy
| | - Giusi Moccia
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy
| | - Ciro Maiello
- Cardiac Transplantation Unit, Department of Cardiac Surgery and Transplantation, Ospedali dei Colli, Naples, Italy
| | - Paride De Rosa
- General Surgery and Transplantation Unit, "San Giovanni di Dio e Ruggi D'Aragona," University Hospital, Scuola Medica Salernitana, Salerno, Italy
| | - Claudio Napoli
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology. Regional Reference Laboratory of Transplant Immunology (LIT) (EFI and ASHI Certifications). Department of Internal Medicine and Specialistics, University of Campania "L. Vanvitelli", Naples, Italy,Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
16
|
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: 31] [Impact Index Per Article: 15.5] [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
|
17
|
Wang L, Ji K, Chen L, Li Y, Zhu W, Yuan X, Bao X, Wu X, He J. Posttransplant de novo DSA and NDSA affect GvHD, OS, and DFS after haplo-HSCT in patients without pre-existing HLA antibodies of hematological malignancies. Front Immunol 2022; 13:1047200. [PMID: 36532004 PMCID: PMC9751004 DOI: 10.3389/fimmu.2022.1047200] [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: 09/17/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022] Open
Abstract
To examine the production time, type, and MFI of post-transplantation de novo HLA antibodies, and their effects on haplo-HSCT outcomes, we retrospectively included 116 patients who were negative for pre-existing HLA antibodies. In total, 322 serum samples from pre-transplantation to post-transplantation were dynamically tested by Luminex and single-antigen bead reagents. Patients were divided into: HLA antibody persistently negative group (group 1), the de novo HLA antibody transiently positive group (group 2), the de novo HLA antibody non-persistently positive group (group 3), and the de novo HLA antibody persistently positive group (group 4). Group 4 included DSA+non-DSA (NDSA) (group 4a) and NDSA (group 4b) groups. The detection rate of de novo HLA antibodies was 75.9% (88/116). The median MFI for de novo HLA antibodies was 2439 (1033-20162). The incidence of II-IV aGvHD was higher in group 2 than in group 1 (52.6% vs 17.9%, P < 0.01); in group 4a than in group 1 (87.5% vs 17.9%, P < 0.001); and in group 4a than in group 4b (87.5% vs 40.0%, P = 0.001). The DFS (37.5% vs 85.7%, P < 0.01) and OS (37.5% vs 85.7%, P < 0.01) of group 4a were lower than those of group 1. The DFS (48.0% vs 85.7%, P < 0.01) and OS (56.0% vs 85.7%, P = 0.03) of group 4b were lower than those of group 1. Multivariate analysis showed that de novo HLA antibody being transiently positive (HR: 5.30; 95% CI: 1.71-16.42, P = 0.01) and persistently positive (HR: 5.67; 95% CI: 2.00-16.08, P < 0.01) were both associated with a higher incidence of II-IV aGvHD. Persistently positive de novo HLA antibodies were a risk factor for reduced DFS (HR: 6.57; 95% CI: 2.08-20.70, P < 0.01) and OS (HR: 5.51; 95% CI: 1.73-17.53, P < 0.01). DSA and NDSA can be detected since 15 days after haplo-HSCT in patients without pre-existing HLA antibodies, and affect aGvHD, DFS, and OS. Haplo-HSCT patients must be monitored for HLA antibodies changes for appropriate preventive clinical management, and we recommend that 1-month post-transplantation is the best test time point.
Collapse
Affiliation(s)
- Lan Wang
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Department of Hematology, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Kai Ji
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Luyao Chen
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying Li
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Department of Hematology, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wenjuan Zhu
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Department of Hematology, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoni Yuan
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaojing Bao
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaojin Wu
- Department of Hematology, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Jun He
- HLA Laboratory of Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China,*Correspondence: Jun He,
| |
Collapse
|
18
|
Rodriguez-Ramirez S, Al Jurdi A, Konvalinka A, Riella LV. Antibody-mediated rejection: prevention, monitoring and treatment dilemmas. Curr Opin Organ Transplant 2022; 27:405-414. [PMID: 35950887 PMCID: PMC9475491 DOI: 10.1097/mot.0000000000001011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Antibody-mediated rejection (AMR) has emerged as the leading cause of late graft loss in kidney transplant recipients. Donor-specific antibodies are an independent risk factor for AMR and graft loss. However, not all donor-specific antibodies are pathogenic. AMR treatment is heterogeneous due to the lack of robust trials to support clinical decisions. This review provides an overview and comments on practical but relevant dilemmas physicians experience in managing kidney transplant recipients with AMR. RECENT FINDINGS Active AMR with donor-specific antibodies may be treated with plasmapheresis, intravenous immunoglobulin and corticosteroids with additional therapies considered on a case-by-case basis. On the contrary, no treatment has been shown to be effective against chronic active AMR. Various biomarkers and prediction models to assess the individual risk of graft failure and response to rejection treatment show promise. SUMMARY The ability to personalize management for a given kidney transplant recipient and identify treatments that will improve their long-term outcome remains a critical unmet need. Earlier identification of AMR with noninvasive biomarkers and prediction models to assess the individual risk of graft failure should be considered. Enrolling patients with AMR in clinical trials to assess novel therapeutic agents is highly encouraged.
Collapse
Affiliation(s)
- Sonia Rodriguez-Ramirez
- Department of Medicine, Division of Nephrology
- Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Ayman Al Jurdi
- Division of Nephrology, Massachusetts General Hospital, Harvard Medical School
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ana Konvalinka
- Department of Medicine, Division of Nephrology
- Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network
- Institute of Medical Science, University of Toronto
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Leonardo V. Riella
- Division of Nephrology, Massachusetts General Hospital, Harvard Medical School
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Lebraud E, Eloudzeri M, Rabant M, Lamarthée B, Anglicheau D. Microvascular Inflammation of the Renal Allograft: A Reappraisal of the Underlying Mechanisms. Front Immunol 2022; 13:864730. [PMID: 35392097 PMCID: PMC8980419 DOI: 10.3389/fimmu.2022.864730] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/22/2022] [Indexed: 12/26/2022] Open
Abstract
Antibody-mediated rejection (ABMR) is associated with poor transplant outcomes and was identified as a leading cause of graft failure after kidney transplantation. Although the hallmark histological features of ABMR (ABMRh), i.e., microvascular inflammation (MVI), usually correlate with the presence of anti-human leukocyte antigen donor-specific antibodies (HLA-DSAs), it is increasingly recognized that kidney transplant recipients can develop ABMRh in the absence of HLA-DSAs. In fact, 40-60% of patients with overt MVI have no circulating HLA-DSAs, suggesting that other mechanisms could be involved. In this review, we provide an update on the current understanding of the different pathogenic processes underpinning MVI. These processes include both antibody-independent and antibody-dependent mechanisms of endothelial injury and ensuing MVI. Specific emphasis is placed on non-HLA antibodies, for which we discuss the ontogeny, putative targets, and mechanisms underlying endothelial toxicity in connection with their clinical impact. A better understanding of these emerging mechanisms of allograft injury and all the effector cells involved in these processes may provide important insights that pave the way for innovative diagnostic tools and highly tailored therapeutic strategies.
Collapse
Affiliation(s)
- Emilie Lebraud
- Necker-Enfants Malades Institute, Inserm U1151, Université de Paris, Department of Nephrology and Kidney Transplantation, Necker Hospital, AP-HP, Paris, France
| | - Maëva Eloudzeri
- Necker-Enfants Malades Institute, Inserm U1151, Université de Paris, Department of Nephrology and Kidney Transplantation, Necker Hospital, AP-HP, Paris, France
| | - Marion Rabant
- Department of Renal Pathology, Necker Hospital, AP-HP, Paris, France
| | - Baptiste Lamarthée
- Université Bourgogne Franche-Comté, EFS BFC, Inserm UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Dany Anglicheau
- Necker-Enfants Malades Institute, Inserm U1151, Université de Paris, Department of Nephrology and Kidney Transplantation, Necker Hospital, AP-HP, Paris, France
| |
Collapse
|
20
|
Karabulut U, Çakır Ü. Non-HDL cholesterol is an independent predictor of long-term cardiovascular events in patients with dyslipidemia after renal transplantation. Int J Clin Pract 2021; 75:e14465. [PMID: 34107128 DOI: 10.1111/ijcp.14465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/19/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Posttransplant dyslipidemia is a common condition in renal transplantation recipients (RTR) and is related to poor cardiac outcomes. We aimed to demonstrate the value of non-high-density lipoprotein cholesterol (non-HDL-C) in predicting long-term major cardiovascular and cerebrovascular events (MACCE) in RTR with dyslipidemia. METHODS Patients who had undergone renal transplantation between 2011 and 2019 were retrospectively analysed and were classified as normal non-HDL-C and high non-HDL-C groups based on first year levels. Development of high non-HDL-C levels was used to predict the occurrence of MACCE (a combination of cardiac death, nonfatal myocardial infarction, unstable angina, and nonfatal stroke) and all-cause death during the long-term follow-up. RESULTS Overall, 674 patients were included, of whom 470 (69.7%) were male; the mean age was 43.6 ± 13.2 years. The mean follow-up duration was 5.5 ± 2.29 years 1 year after the transplant. MACCE occurred during the follow-up in 102 (61.8%) patients in the high non-HDL-C group and 13 (2.6%) patients in the normal non-HDL-C group (P < .001). High non-HDL-C was a predictor of MACCE in the multivariate analysis (hazard ratio [HR] 1.02, 95% confidence interval [CI] 1.01-1.02, P < .001). Smoking (HR: 1.92, 95% CI 1.16-3.20, P < .001), cadaver graft (HR: 2.55, 95% CI 1.52-4.26, P < .001), and left ventricular ejection fraction (HR: 0.96, 95% CI 0.94-0.98, P < .001) were also predictors of MACCE. Kaplan-Meier analysis revealed that all MACCE components and all-cause mortality were significantly higher in the high non-HDL-C group (P < .001). CONCLUSION Non-HDL-C was closely related to long-term cardiac outcomes in RTR with dyslipidemia. Non-HDL-C should be among the primary goals in lipid-lowering treatment in post-transplant dyslipidemia.
Collapse
Affiliation(s)
- Umut Karabulut
- Department of Cardiology, Acıbadem International Hospital, İstanbul, Turkey
| | - Ülkem Çakır
- Department of Nephrology, Facult of Medicine, Acıbadem University, İstanbul, Turkey
| |
Collapse
|
21
|
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
|
22
|
Shimabukuro S, Iwasaki K, Kawai S, Shirouzu T, Miwa Y, Iida Y, Nakajima F, Horimi K, Matsuoka Y, Ashimine S, Ishiyama K, Kobayashi T. Improved detection of donor-specific HLA-class II antibody in kidney transplant recipients by modified immunocomplex capture fluorescence analysis. Transpl Immunol 2021; 67:101418. [PMID: 34052300 DOI: 10.1016/j.trim.2021.101418] [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: 04/16/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022]
Abstract
Immunocomplex capture fluorescence analysis (ICFA) which basic principle is same as Luminex crossmatch (LXM), could detect donor-specific HLA antibody (DSA). The advantages of ICFA are (i) detection of DSA and (ii) no requirement of viable cells over the flow cytometry crossmatch (FCXM). However, FCXM has been widely used because of its higher sensitivity than ICFA, in particular HLA-class II antibody detection. In this study the accuracy of DSA detection against HLA-class II was investigated by modifying the original method of ICFA. Increment of the sensitivity was found when purified peripheral blood mononuclear cells (PBMCs) were used instead of whole blood. An ICFA-PBMC in addition to FCXM-T/B was conducted for 118 patients before kidney transplantation and 13 patients with de novo DSA against HLA-class II after transplantation. Significantly positive correlation was observed between the values of ICFA-PBMC and DSA mean fluorescence intensity (MFI) targeting class II (p < 0.0001). When the cutoff level of 1.4 was determined by receiver operating characteristic curve analysis, the average DSA MFI was found to be significantly higher in the ICFA-PBMC (class II) positive group comparing to that in the negative group (12,217 vs 3885, p = 0.0027). ICFA-PBMC and optimized cutoff level could provide valid information in cases of suspected DSA.
Collapse
Affiliation(s)
- Shuichi Shimabukuro
- Department of Urology, Okinawa Chubu Hospital, 281, Miyazato, Uruma, Okinawa, Japan; Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenta Iwasaki
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan.
| | - Shintaro Kawai
- Wakunaga Pharmaceutical Co, Ltd, Molecular Diagnostics Division, Osaka, Japan
| | - Takayuki Shirouzu
- Wakunaga Pharmaceutical Co, Ltd, Molecular Diagnostics Division, Osaka, Japan
| | - Yuko Miwa
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yusuke Iida
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Fumiaki Nakajima
- Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Kosei Horimi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yutaka Matsuoka
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Satoshi Ashimine
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kohei Ishiyama
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
23
|
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
|
24
|
Tomita Y, Ishida H, Uehara S, Takiguchi S, Sato T, Nakamura M. CD45RA -CD25 highCD127 -CD4 + activated regulatory T cells are correlated with de novo donor-specific anti-HLA antibody formation after kidney transplantation in standard immunosuppression. Int Immunopharmacol 2021; 97:107661. [PMID: 33887579 DOI: 10.1016/j.intimp.2021.107661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/22/2021] [Accepted: 04/05/2021] [Indexed: 12/16/2022]
Abstract
Although de novo donor-specific anti-HLA antibodies (dnDSA) remain a barrier for human kidney transplantation (KTx), the role of regulatory T (Treg) cells in dnDSA formation remains unknown. To address this question, we evaluated Treg cell subsets in peripheral blood mononuclear cells in 15 healthy volunteers and 59 KTx recipients using flow cytometric analysis. The post-transplant CD25highCD127-CD4+ Treg cells in KTx recipients were down-regulated compared with those of healthy volunteers (P < .001). Among them, 11 KTx recipients showed dnDSA formation, which was associated with lower frequencies of CD25highCD127-CD4+ Treg cells (P = .040). Furthermore, of the total Treg cell population, CD45RA-CD25highCD127-CD4+ activated Treg (aTreg) cells were significantly dominant in patients with dnDSA (P = .038), but not CD45RA+CD25highCD127-CD4+ resting Treg cells (P = .961). In contrast, non-donor-specific anti-HLA antibody formation was not associated with CD45RA- aTreg cells (P = .772). Multivariate logistic regression analyses revealed that CD45RA- aTreg cells were independently associated with dnDSA formation (Odds ratio = 6.69, P = .040). These findings indicate that CD45RA- aTreg cells are strongly associated with dnDSA formation in KTx recipients and might be an important risk factor of antibody-mediated rejection before clinical diagnosis.
Collapse
Affiliation(s)
- Yusuke Tomita
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan.
| | - Hiroaki Ishida
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Saeko Uehara
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Shinya Takiguchi
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Takehito Sato
- Department of Immunology, Tokai University School of Medicine, Kanagawa, Japan
| | - Michio Nakamura
- Department of Transplant Surgery, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
25
|
Blydt-Hansen TD, Sharma A, Gibson IW, Wiebe C, Sharma AP, Langlois V, Teoh CW, Rush D, Nickerson P, Wishart D, Ho J. Validity and utility of urinary CXCL10/Cr immune monitoring in pediatric kidney transplant recipients. Am J Transplant 2021; 21:1545-1555. [PMID: 33034126 DOI: 10.1111/ajt.16336] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/26/2020] [Accepted: 09/20/2020] [Indexed: 02/06/2023]
Abstract
Individualized posttransplant immunosuppression is hampered by suboptimal monitoring strategies. To validate the utility of urinary CXCL10/Cr immune monitoring in children, we conducted a multicenter prospective observational study in children <21 years with serial and biopsy-associated urine samples (n = 97). Biopsies (n = 240) were categorized as normal (NOR), rejection (>i1t1; REJ), indeterminate (IND), BKV infection, and leukocyturia (LEU). An independent pediatric cohort of 180 urines was used for external validation. Ninety-seven patients aged 11.4 ± 5.5 years showed elevated urinary CXCL10/Cr in REJ (3.1, IQR 1.1, 16.4; P < .001) and BKV nephropathy (median = 5.6, IQR 1.3, 26.9; P < .001) vs. NOR (0.8, IQR 0.4, 1.5). The AUC for REJ vs. NOR was 0.76 (95% CI 0.66-0.86). Low (0.63) and high (4.08) CXCL10/Cr levels defined high sensitivity and specificity thresholds, respectively; validated against an independent sample set (AUC = 0.76, 95% CI 0.66-0.86). Serial urines anticipated REJ up to 4 weeks prior to biopsy and declined within 1 month following treatment. Elevated mean CXCL10/Cr was correlated with first-year eGFR decline (ρ = -0.37, P ≤ .001), particularly when persistently exceeding ≥4.08 (ratio = 0.81; P < .04). Useful thresholds for urinary CXCL10/Cr levels reproducibly define the risk of rejection, immune quiescence, and decline in allograft function for use in real-time clinical monitoring in children.
Collapse
Affiliation(s)
- Tom D Blydt-Hansen
- Pediatric Nephrology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Atul Sharma
- Biostatistical Consulting Unit, George, Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ian W Gibson
- Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chris Wiebe
- Nephrology, University of Manitoba, Winnipeg, Manitoba, Canada.,Transplant/Immunology Lab, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ajay P Sharma
- Pediatric Nephrology, University of Western Ontario, London, Ontario, Canada
| | - Valerie Langlois
- Pediatric Nephrology, University of Toronto, Toronto, Ontario, Canada
| | - Chia W Teoh
- Pediatric Nephrology, University of Toronto, Toronto, Ontario, Canada
| | - David Rush
- Nephrology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter Nickerson
- Nephrology, University of Manitoba, Winnipeg, Manitoba, Canada.,Transplant/Immunology Lab, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Wishart
- Computing Science, University of Alberta, Edmonton, Alberta, Canada.,The Metabolomics Innovation Center, Edmonton, Alberta, Canada
| | - Julie Ho
- Nephrology, University of Manitoba, Winnipeg, Manitoba, Canada.,Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
| |
Collapse
|
26
|
Gautier Vargas G, Olagne J, Parissiadis A, Joly M, Cognard N, Perrin P, Froelich N, Guntz P, Gachet C, Moulin B, Caillard S. Does a Useful Test Exist to Properly Evaluate the Pathogenicity of Donor-specific Antibodies? Lessons From a Comprehensive Analysis in a Well-studied Single-center Kidney Transplant Cohort. Transplantation 2021; 104:2148-2157. [PMID: 31895344 DOI: 10.1097/tp.0000000000003080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Donor-specific antibodies (DSA) play a major role in antibody-mediated rejection (AMR) and graft dysfunction. However, the clinical relevance of complement-binding anti-HLA antibodies remains unclear. METHODS Here, we analyzed DSA detected in the serum (sDSA) using single antigen bead, C1q, and C3d assays combined with the study of intragraft DSA (gDSA) in 86 patients who had DSA and underwent a kidney biopsy for cause (n = 58) or without evidence of kidney dysfunction (n = 28). DSA characteristics were collected and related to the presence of AMR, graft histological features, and allograft survival. RESULTS Forty-five patients (52%) had C1q DSA, and 42 (51%) had C3d DSA. Allograft biopsies revealed AMR in 63 cases (73%), regardless of kidney function. gDSA were identified in 74% of biopsies. We observed a strong correlation among single antigen bead mean fluorescence intensity and complement assays positivity, presence of gDSA, and AMR occurrence. CONCLUSIONS Complement-binding DSA per se were not significantly associated with allograft survival in the entire study sample. Finally, gDSA predicted subsequent graft loss in patients who showed a stable renal function at the day of biopsy. Our data suggest that DSA mean fluorescence intensity and presence of gDSA might provide prognostic information during posttransplant monitoring.
Collapse
Affiliation(s)
| | - Jérome Olagne
- Nephrology-Transplantation Department, University Hospital, Strasbourg, France.,Department of Pathology, University Hospital, Strasbourg, France
| | - Anne Parissiadis
- Histocompatibility Laboratory, UMR-S949 Inserm, Etablissement Français du Sang, Strasbourg, France
| | - Mélanie Joly
- Nephrology-Transplantation Department, University Hospital, Strasbourg, France
| | - Noelle Cognard
- Nephrology-Transplantation Department, University Hospital, Strasbourg, France
| | - Peggy Perrin
- Nephrology-Transplantation Department, University Hospital, Strasbourg, France
| | - Nadine Froelich
- Histocompatibility Laboratory, UMR-S949 Inserm, Etablissement Français du Sang, Strasbourg, France
| | - Philippe Guntz
- Histocompatibility Laboratory, UMR-S949 Inserm, Etablissement Français du Sang, Strasbourg, France
| | - Christian Gachet
- Histocompatibility Laboratory, UMR-S949 Inserm, Etablissement Français du Sang, Strasbourg, France
| | - Bruno Moulin
- Nephrology-Transplantation Department, University Hospital, Strasbourg, France.,INSERM UMR_S 1109, ImmunoRhumatologie Moléculaire, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Institut d'Immunologie et d'Hématologie, Strasbourg, France
| | - Sophie Caillard
- Nephrology-Transplantation Department, University Hospital, Strasbourg, France.,INSERM UMR_S 1109, ImmunoRhumatologie Moléculaire, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Institut d'Immunologie et d'Hématologie, Strasbourg, France
| |
Collapse
|
27
|
Hiramitsu T, Tomosugi T, Futamura K, Okada M, Nishihira M, Goto N, Ichimori T, Narumi S, Kobayashi T, Uchida K, Watarai Y. Optimal blood levels of (extended-release) tacrolimus in living donor kidney transplantation to prevent de novo donor-specific antibody production: A retrospective cohort study. Int Immunopharmacol 2020; 91:107038. [PMID: 33388731 DOI: 10.1016/j.intimp.2020.107038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/03/2020] [Accepted: 09/21/2020] [Indexed: 01/25/2023]
Abstract
Chronic antibody-mediated rejection, caused by de novo donor-specific antibody (dnDSA) production, results in poor graft survival. To prevent dnDSA production, optimal blood levels of immunosuppressive drugs in living donor kidney transplant recipients were determined. A total of 772 recipients underwent living donor kidney transplantation between January 2008 and December 2017. Graft survival and risk factors for dnDSA production were investigated in 647 recipients. Optimal blood levels of tacrolimus (TAC) and extended-release TAC (TACER) were measured in recipients receiving steroids and mycophenolate mofetil, combined with TAC (n = 53) or TACER (n = 135). Receiver operating characteristic (ROC) curve analysis and comparisons between dnDSA-negative and dnDSA-positive recipients were carried out. The Kaplan-Meier method revealed significantly poor graft survival in dnDSA-positive recipients (P < 0.001). Cox regression models indicated calcineurin inhibitor withdrawal as a significant risk for dnDSA production (P < 0.001; hazard ratio 6.637; 95% confidence interval 2.667-6.517). Average trough levels of TAC and TACER in dnDSA-negative recipients were significantly higher than those in dnDSA-positive recipients (4.88 vs 3.69 ng TAC/ml, P = 0.023, and 4.60 vs 3.85 ng TACER/ml, P = 0.001). ROC curve analysis indicated 4.325 and 3.990 ng/ml as the best trough levels under TAC- and TACER-based regimens, respectively, to prevent dnDSA production (areas under the curve: 0.788 and 0.813, respectively). Maintenance of the trough levels of TAC > 4.325 ng/ml and TACER > 3.990 ng/ml may prevent dnDSA production.
Collapse
Affiliation(s)
- Takahisa Hiramitsu
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan.
| | - Toshihide Tomosugi
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Kenta Futamura
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Manabu Okada
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Morikuni Nishihira
- Department of Renal Transplant Surgery, Masuko Memorial Hospital, 453-8566 35-28 Takehashi-cho, Nakamura-ku, Nagoya, Aichi, Japan
| | - Norihiko Goto
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Toshihiro Ichimori
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Shunji Narumi
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, 480-1195 1-1 Yazakokarimata, Nagakute, Aichi, Japan
| | - Kazuharu Uchida
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan; Department of Renal Transplant Surgery, Masuko Memorial Hospital, 453-8566 35-28 Takehashi-cho, Nakamura-ku, Nagoya, Aichi, Japan
| | - Yoshihiko Watarai
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross Hospital, 466-8650 2-9 Myoken-cho, Showa-ku, Nagoya, Aichi, Japan
| |
Collapse
|
28
|
Safety and Efficacy of a Steroid Avoidance Immunosuppression Regimen in Renal Transplant Patients With De Novo or Preformed Donor-Specific Antibodies: A Single-Center Study. Transplant Proc 2020; 53:950-961. [PMID: 33293041 DOI: 10.1016/j.transproceed.2020.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 11/23/2022]
Abstract
Although interest in the role of donor-specific antibodies (DSAs) in kidney transplant rejection, graft survival, and histopathological outcomes is increasing, their impact on steroid avoidance or minimization in renal transplant populations is poorly understood. Primary outcomes of graft survival, rejection, and histopathological findings were assessed in 188 patients who received transplants between 2012 and 2015 at the Scripps Center for Organ Transplantation, which follows a steroid avoidance protocol. Analyses were performed using data from the United Network for Organ Sharing. Cohorts included kidney transplant recipients with de novo DSAs (dnDSAs; n = 27), preformed DSAs (pfDSAs; n = 15), and no DSAs (nDSAs; n = 146). Median time to dnDSA development (classes I and II) was shorter (102 days) than in previous studies. Rejection of any type was associated with DSAs to class I HLA (P < .05) and class II HLA (P < .01) but not with graft loss. Although mean fluorescence intensity (MFI) independently showed no association with rejection, an MFI >5000 showed a trend toward more antibody-mediated rejection (P < .06), though graft loss was not independently associated. Banff chronic allograft nephropathy scores and a modified chronic injury score were increased in the dnDSA cohort at 6 months, but not at 2 years (P < .001 and P < .08, respectively). Our data suggest that dnDSAs and pfDSAs impact short-term rejection rates but do not negatively impact graft survival or histopathological outcomes at 2 years. Periodic protocol post-transplant DSA monitoring may preemptively identify patients who develop dnDSAs who are at a higher risk for rejection.
Collapse
|
29
|
Protocol Biopsies in Patients With Subclinical De Novo Donor-specific Antibodies After Kidney Transplantation: A Multicentric Study. Transplantation 2020; 104:1726-1737. [PMID: 32732853 DOI: 10.1097/tp.0000000000003055] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND De novo donor-specific antibodies (DSAs) are associated with antibody-mediated rejection (AMR) and allograft loss. Whether monitoring of de novo DSA (dnDSA) paired with systematic kidney biopsy should become routine remains to be established. METHODS A retrospective multicentric study (9 French kidney transplant units of the Spiesser group) included patients without graft dysfunction biopsied because of the presence of dnDSA (One Lambda, mean fluorescence intensity [MFI], >1000). RESULTS One hundred twenty-three patients (85 male/38 female; mean age, 49.5 ± 13.1 y old) were biopsied after the detection of a dnDSA, 65.3 months (median) after kidney transplantation. Graft function was stable within 3 months before biopsy (estimated glomerular filtration rate, 55.3 ± 18.9 mL/min/1.73 m). Fifty-one subclinical AMRs (sAMRs) (41.4%) were diagnosed, of which 32 (26%) active and 19 (15.5%) chronic active sAMR. Seventy-two biopsies revealed no AMR (58.5%). Predictive factors associated with the diagnosis of active sAMR were MFI of immunodominant DSA >4000, MFI of the sum of DSA >6300, age of the recipient <45 years old, and the absence of steroids at biopsy. The presence of proteinuria >200 mg/g was predictive of chronic active sAMR. The decrease of estimated glomerular filtration rate at 5 years post-biopsy was significantly higher in patients with acute sAMR (-25.2 ± 28.3 mL/min/1.73 m) and graft survival significantly lower. CONCLUSIONS Performing a kidney graft biopsy for the occurrence of dnDSA without renal dysfunction leads to the diagnosis of a sAMR in over 40% of cases. Nevertheless, we did not observe any effect of standard treatment in acute sAMR.
Collapse
|
30
|
Cordero E, Bulnes-Ramos A, Aguilar-Guisado M, González Escribano F, Olivas I, Torre-Cisneros J, Gavaldá J, Aydillo T, Moreno A, Montejo M, Fariñas MC, Carratalá J, Muñoz P, Blanes M, Fortún J, Suárez-Benjumea A, López-Medrano F, Roca C, Lara R, Pérez-Romero P. Effect of Influenza Vaccination Inducing Antibody Mediated Rejection in Solid Organ Transplant Recipients. Front Immunol 2020; 11:1917. [PMID: 33123119 PMCID: PMC7574595 DOI: 10.3389/fimmu.2020.01917] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Our goal was to study whether influenza vaccination induced antibody mediated rejection in a large cohort of solid organ transplant recipients (SOTR). Methods Serum anti-Human Leukocyte Antigen (HLA) antibodies were determined using class I and class II antibody-coated latex beads (FlowPRATM Screening Test) by flow cytometry. Anti-HLA antibody specificity was determined using the single-antigen bead flow cytometry (SAFC) assay and assignation of donor specific antibodies (DSA) was performed by virtual-crossmatch. Results We studied a cohort of 490 SOTR that received an influenza vaccination from 2009 to 2013: 110 (22.4%) received the pandemic adjuvanted vaccine, 59 (12%) within the first 6 months post-transplantation, 185 (37.7%) more than 6 months after transplantation and 136 (27.7%) received two vaccination doses. Overall, no differences of anti-HLA antibodies were found after immunization in patients that received the adjuvanted vaccine, within the first 6 months post-transplantation, or based on the type of organ transplanted. However, the second immunization dose increased the percentage of patients positive for anti-HLA class I significantly compared with patients with one dose (14.6% vs. 3.8%; P = 0.003). Patients with pre-existing antibodies before vaccination (15.7% for anti-HLA class I and 15.9% for class II) did not increase reactivity after immunization. A group of 75 (14.4%) patients developed de novo anti-HLA antibodies, however, only 5 (1.02%) of them were DSA, and none experienced allograft rejection. Only two (0.4%) patients were diagnosed with graft rejection with favorable outcomes and neither of them developed DSA. Conclusion Our results suggest that influenza vaccination is not associated with graft rejection in this cohort of SOTR.
Collapse
Affiliation(s)
- Elisa Cordero
- Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain.,Department of Medicine, University of Seville, Seville, Spain
| | - Angel Bulnes-Ramos
- Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | - Manuela Aguilar-Guisado
- Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | - Francisca González Escribano
- Servicio de Inmunología, Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | - Israel Olivas
- Servicio de Inmunología, Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | - Julián Torre-Cisneros
- Reina Sofia University Hospital, Maimonides Institute for Biomedical Research (IMIBIC), University of Córdoba (UCO), Córdoba, Spain
| | - Joan Gavaldá
- Vall d'Hebron University Hospital, VHIR, Barcelona, Spain
| | - Teresa Aydillo
- Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | | | | | | | - Jordi Carratalá
- Belltvitge University Hospital, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigaciónn Biomédica Gregorio Marañón, Madrid, Spain.,Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,CIBERES (CB06/06/0058), Madrid, Spain
| | | | - Jesús Fortún
- University Hospital Ramón y Cajal, Madrid, Spain
| | | | - Francisco López-Medrano
- Unit of Infectious Diseases, University Hospital 12 de Octubre, Madrid, Spain.,Instituto de Investigación Biomédica imas12, Madrid, Spain.,Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Roca
- Instituto de Biomedicina de Sevilla (IBIS), University Hospital Virgen del Rocío, CSIC, University of Seville, Seville, Spain
| | - Rosario Lara
- Reina Sofia University Hospital, Maimonides Institute for Biomedical Research (IMIBIC), University of Córdoba (UCO), Córdoba, Spain
| | - Pilar Pérez-Romero
- National Centre for Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
31
|
Wiebe C, Rush DN, Gibson IW, Pochinco D, Birk PE, Goldberg A, Blydt‐Hansen T, Karpinski M, Shaw J, Ho J, Nickerson PW. Evidence for the alloimmune basis and prognostic significance of Borderline T cell-mediated rejection. Am J Transplant 2020; 20:2499-2508. [PMID: 32185878 PMCID: PMC7496654 DOI: 10.1111/ajt.15860] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/05/2020] [Accepted: 03/07/2020] [Indexed: 02/06/2023]
Abstract
Prognostic biomarkers of T cell-mediated rejection (TCMR) have not been adequately studied in the modern era. We evaluated 803 renal transplant recipients and correlated HLA-DR/DQ molecular mismatch alloimmune risk categories (low, intermediate, high) with the severity, frequency, and persistence of TCMR. Allograft survival was reduced in recipients with Banff Borderline (hazard ratio [HR] 2.4, P = .003) and Banff ≥ IA TCMR (HR 4.3, P < .0001) including a subset who never developed de novo donor-specific antibodies (P = .002). HLA-DR/DQ molecular mismatch alloimmune risk categories were multivariate correlates of Banff Borderline and Banff ≥ IA TCMR and correlated with the severity and frequency of rejection episodes. Recipient age, HLA-DR/DQ molecular mismatch category, and cyclosporin vs tacrolimus immunosuppression were independent correlates of Banff Borderline and Banff ≥ IA TCMR. In the subset treated with tacrolimus (720/803) recipient age, HLA-DR/DQ molecular mismatch category, and tacrolimus coefficient of variation were independent correlates of TCMR. The correlation of HLA-DR/DQ molecular mismatch category with TCMR, including Borderline, provides evidence for their alloimmune basis. HLA-DR/DQ molecular mismatch may represent a precise prognostic biomarker that can be applied to tailor immunosuppression or design clinical trials based on individual patient risk.
Collapse
Affiliation(s)
- Chris Wiebe
- Department of MedicineUniversity of ManitobaWinnipegCanada
- Shared Health Services ManitobaWinnipegCanada
- Department of ImmunologyUniversity of ManitobaWinnipegCanada
| | - David N. Rush
- Department of MedicineUniversity of ManitobaWinnipegCanada
| | - Ian W. Gibson
- Shared Health Services ManitobaWinnipegCanada
- Department of PathologyUniversity of ManitobaWinnipegCanada
| | | | - Patricia E. Birk
- Department of Pediatrics and Child HealthUniversity of ManitobaWinnipegCanada
| | - Aviva Goldberg
- Department of Pediatrics and Child HealthUniversity of ManitobaWinnipegCanada
| | - Tom Blydt‐Hansen
- Department of PediatricsUniversity of British ColumbiaWinnipegCanada
| | | | - Jamie Shaw
- Department of MedicineUniversity of ManitobaWinnipegCanada
| | - Julie Ho
- Department of MedicineUniversity of ManitobaWinnipegCanada
- Department of ImmunologyUniversity of ManitobaWinnipegCanada
| | - Peter W. Nickerson
- Department of MedicineUniversity of ManitobaWinnipegCanada
- Shared Health Services ManitobaWinnipegCanada
- Department of ImmunologyUniversity of ManitobaWinnipegCanada
| |
Collapse
|
32
|
Sakamoto S, Iwasaki K, Tomosugi T, Niemann M, Spierings E, Miwa Y, Horimi K, Takeda A, Goto N, Narumi S, Watarai Y, Kobayashi T. Analysis of T and B Cell Epitopes to Predict the Risk of de novo Donor-Specific Antibody (DSA) Production After Kidney Transplantation: A Two-Center Retrospective Cohort Study. Front Immunol 2020; 11:2000. [PMID: 32973806 PMCID: PMC7481442 DOI: 10.3389/fimmu.2020.02000] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/23/2020] [Indexed: 01/25/2023] Open
Abstract
Risk prediction of de novo donor specific antibody (DSA) would be very important for long term graft outcome after organ transplantation. The purpose of this study was to elucidate the association of eplet mismatches and predicted indirectly recognizable HLA epitopes (PIRCHE) scores with de novo DSA production. Our retrospective cohort study enrolled 691 living donor kidney transplantations. HLA-A, B, DRB and DQB eplet mismatches and PIRCHE scores (4 digit of HLA-A, B, DR, and DQ) were determined by HLA matchmaker (ver 2.1) and PIRCHE-II Matching Service, respectively. Weak correlation between eplet mismatches and PIRCHE scores was identified, although both measurements were associated with classical HLA mismatches. Class II (DRB+DQB) eplet mismatches were significantly correlated with the incidence of de novo class II (DR/DQ) DSA production [8/235 (3.4%) in eplet mismatch ≤ 13 vs. 92/456 (20.2%) in eplet mismatch ≥ 14, p < 0.001]. PIRCHE scores were also significantly correlated with de novo class II DSA production [26/318 (8.2%) in PIRCHE ≤ 175 vs. 74/373 (19.8%) in PIRCHE ≥ 176, p < 0.001]. Patients with low levels of both class II eplet mismatches and PIRCHE scores developed de novo class II DSA only in 4/179 (2.2%). Analysis of T cell and B cell epitopes can provide a beneficial information on the design of individualized immunosuppression regimens for prevention of de novo DSA production after kidney transplantation.
Collapse
Affiliation(s)
- Shintaro Sakamoto
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan.,Department of Histocompatibility Laboratory, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Kenta Iwasaki
- Department of Kidney Diseases and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Toshihide Tomosugi
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | | | - Eric Spierings
- Laboratory of Translational Immunology, UMC Utrecht, Utrecht, Netherlands
| | - Yuko Miwa
- Department of Kidney Diseases and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kosei Horimi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Asami Takeda
- Department of Nephrology, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Norihiko Goto
- Department of Transplant Internal Medicine, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Shunji Narumi
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Yoshihiko Watarai
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
33
|
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
|
34
|
Iwasaki K, Hamana H, Kishi H, Yamamoto T, Hiramitsu T, Okad M, Tomosugi T, Takeda A, Narumi S, Watarai Y, Miwa Y, Okumura M, Matsuoka Y, Horimi K, Muraguchi A, Kobayash T. The suppressive effect on CD4 T cell alloresponse against endothelial HLA-DR via PD-L1 induced by anti-A/B ligation. Clin Exp Immunol 2020; 202:249-261. [PMID: 32578199 DOI: 10.1111/cei.13482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 06/15/2020] [Indexed: 11/27/2022] Open
Abstract
While donor-specific human leukocyte antigen (HLA) antibodies are a frequent cause for chronic antibody-mediated rejection in organ transplantation, this is not the case for antibodies targeting blood group antigens, as ABO-incompatible (ABO-I) organ transplantation has been associated with a favorable graft outcome. Here, we explored the role of CD4 T cell-mediated alloresponses against endothelial HLA-D-related (DR) in the presence of anti-HLA class I or anti-A/B antibodies. CD4 T cells, notably CD45RA-memory CD4 T cells, undergo extensive proliferation in response to endothelial HLA-DR. The CD4 T cell proliferative response was enhanced in the presence of anti-HLA class I, but attenuated in the presence of anti-A/B antibodies. Microarray analysis and molecular profiling demonstrated that the expression of CD274 programmed cell death ligand 1 (PD-L1) increased in response to anti-A/B ligation-mediated extracellular signal-regulated kinase (ERK) inactivation in endothelial cells that were detected even in the presence of interferon-γ stimulation. Anti-PD-1 antibody enhanced CD4 T cell proliferation, and blocked the suppressive effect of the anti-A/B antibodies. Educated CD25+ CD127- regulatory T cells (edu.Tregs ) were more effective at preventing CD4 T cell alloresponses to endothelial cells compared with naive Treg ; anti-A/B antibodies were not involved in the Treg -mediated events. Finally, amplified expression of transcript encoding PD-L1 was observed in biopsy samples from ABO-I renal transplants when compared with those from ABO-identical/compatible transplants. Taken together, our findings identified a possible factor that might prevent graft rejection and thus contribute to a favorable outcome in ABO-I renal transplantation.
Collapse
Affiliation(s)
- K Iwasaki
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - H Hamana
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - H Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - T Yamamoto
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - T Hiramitsu
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - M Okad
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - T Tomosugi
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - A Takeda
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - S Narumi
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Y Watarai
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Y Miwa
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - M Okumura
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Y Matsuoka
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - K Horimi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - A Muraguchi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - T Kobayash
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| |
Collapse
|
35
|
Geoghegan L, Al-Khalil M, Scarborough A, Murray A, Issa F. Pre-transplant management and sensitisation in vascularised composite allotransplantation: A systematic review. J Plast Reconstr Aesthet Surg 2020; 73:1593-1603. [PMID: 32475735 DOI: 10.1016/j.bjps.2020.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 04/11/2020] [Accepted: 05/09/2020] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Vascularised composite allotransplantation (VCA) permits like-for-like reconstruction following extensive soft tissue injuries. The initial management of extensive soft tissue injury can lead to the development of anti-HLA antibodies through injury-related factors, transfusion and cadaveric grafting. The role of antibody-mediated rejection, donor-specific antibody formation and graft rejection in the context of VCA remains unclear. This systematic review aimed to determine whether pre-transplant management strategies influence immunological outcome following VCA. METHODS A systematic review of MEDLINE, EMBASE and CINAHL using a PRISMA-compliant methodology up to February 2019 was conducted. Pre-transplant, procedural and long-term outcome data were collected and recorded for all VCA recipients on an individual patient basis. RESULTS The search revealed 3,847 records of which 114 met inclusion criteria and reported clinical data related to 100 patients who underwent 129 VCA transplants. Trauma (50%) and burns (15%) were the most frequent indications for VCA. Of all 114 studies, only one reported acute resuscitative management. Fifteen patients were sensitised prior to reconstructive transplantation with an 80%%incidence of acute rejection in the first post-operative year. Seven patients demonstrated graft vasculopathy, only one of whom had demonstrated panel reactive antibodies. CONCLUSIONS Currently employed acute management strategies may predispose to the development of anti-HLA antibodies, adding to the already complex immunological challenge of VCA. To determine whether association between pre-transplant management and outcomes exists, further refinement of international registries is required.
Collapse
Affiliation(s)
- Luke Geoghegan
- Imperial College NHS Trust, London, UK; Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science (NDORMS), University of Oxford, Oxford, UK
| | | | | | - Alexandra Murray
- Department of Plastic and Reconstructive Surgery, Stoke Mandeville Hospital, Aylesbury, UK
| | - Fadi Issa
- Department of Plastic and Reconstructive Surgery, Stoke Mandeville Hospital, Aylesbury, UK; Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
36
|
Usureau C, Jacob V, Clichet V, Presne C, Desoutter J, Poulain C, Choukroun G, Guillaume N. Flow cytometry crossmatching to investigate kidney-biopsy-proven, antibody-mediated rejection in patients who develop de novo donor-specific antibodies. Transpl Immunol 2020; 61:101303. [PMID: 32387224 DOI: 10.1016/j.trim.2020.101303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The appearance of de novo donor-specific anti-human leukocyte antigen antibodies (dnDSAs) after kidney transplantation is independently associated with poor long-term allograft outcomes. The objective of the present study was to evaluate the predictive value of a flow cytometry crossmatching (FC-XM) assay after the appearance of dnDSAs related to antibody-mediated allograft rejection (ABMR) after kidney transplantation. MATERIALS AND METHODS A total of 89 recipients with dnDSAs after transplantation were included. The crossmatching results were compared with the dnDSA profile (the mean fluorescence intensity (MFI), the complement-binding activity, and the IgG subclass profile) and the biopsy's morphological features. RESULTS Of the 89 patients, 59 (66%) were positive in an FC-XM assay, 17 (19%) had complement-binding DSAs, 55 (62%) were positive for IgG1 and/or IgG3 in a solid phase assay, and 45 (51%) had morphological biopsy features linked to ABMR. CONCLUSION An FC-XM assay was unable to discriminate between cases with or without ABMR on biopsy findings; it had a low positive predictive value (<70%) and a low negative positive predictive value (<42.9%), taking into account the sensitivity of our assay (limit of detection: DSAs with an MFI >3000). In this context, the height of the MFI of the dnDSAs might be enough for a high positive predictive value for ABMR and additional testing for complement binding activity can remain optional.
Collapse
Affiliation(s)
- Cédric Usureau
- Department of Haematology and Histocompatibility, Amiens University Hospital, Amiens, France; EA Hematim, Jules Verne University of Picardie, Amiens, France
| | - Valentine Jacob
- Department of Haematology and Histocompatibility, Amiens University Hospital, Amiens, France; EA Hematim, Jules Verne University of Picardie, Amiens, France
| | - Valentin Clichet
- Department of Haematology and Histocompatibility, Amiens University Hospital, Amiens, France; EA Hematim, Jules Verne University of Picardie, Amiens, France
| | - Claire Presne
- Department of Nephrology and Transplantation, Amiens University Hospital, Amiens, France
| | - Judith Desoutter
- Department of Haematology and Histocompatibility, Amiens University Hospital, Amiens, France; EA Hematim, Jules Verne University of Picardie, Amiens, France
| | - Coralie Poulain
- Department of Nephrology and Transplantation, Amiens University Hospital, Amiens, France
| | - Gabriel Choukroun
- Department of Nephrology and Transplantation, Amiens University Hospital, Amiens, France
| | - Nicolas Guillaume
- Department of Haematology and Histocompatibility, Amiens University Hospital, Amiens, France; EA Hematim, Jules Verne University of Picardie, Amiens, France.
| |
Collapse
|
37
|
Abstract
Early detection of graft injury after kidney transplantation is key to maintaining long-term good graft function. Graft injury could be due to a multitude of factors including ischaemia reperfusion injury, cell or antibody-mediated rejection, progressive interstitial fibrosis and tubular atrophy, infections and toxicity from the immunosuppressive drugs themselves. The current gold standard for assessing renal graft dysfunction is renal biopsy. However, biopsy is usually late when triggered by a change in serum creatinine and of limited utility in diagnosis of early injury when histological changes are equivocal. Therefore, there is a need for timely, objective and non-invasive diagnostic techniques with good early predictive value to determine graft injury and provide precision in titrating immunosuppression. We review potential novel plasma and urine biomarkers that offer sensitive new strategies for early detection and provide major insights into mechanisms of graft injury. This is a rapidly expanding field, but it is likely that a combination of biomarkers will be required to provide adequate sensitivity and specificity for detecting graft injury.
Collapse
|
38
|
Maenaka A, Kenta I, Ota A, Miwa Y, Ohashi W, Horimi K, Matsuoka Y, Ohnishi M, Uchida K, Kobayashi T. Interferon-γ-induced HLA Class II expression on endothelial cells is decreased by inhibition of mTOR and HMG-CoA reductase. FEBS Open Bio 2020; 10:927-936. [PMID: 32237049 PMCID: PMC7193171 DOI: 10.1002/2211-5463.12854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/10/2020] [Accepted: 03/26/2020] [Indexed: 12/15/2022] Open
Abstract
In organ transplantation, donor‐specific HLA antibody (DSA) is considered a major cause of graft rejection. Because DSA targets primarily donor‐specific human leukocyte antigen (HLA) expressed on graft endothelial cells, the prevention of its expression is a possible strategy for avoiding or salvaging DSA‐mediated graft rejection. We examined the effect of various clinically used drugs on HLA class II expression on endothelial cells. Interferon‐γ (IFN‐γ)‐induced HLA class II DR (HLA‐DR) was downregulated by everolimus (EVR, 49.1% ± 0.8%; P < 0.01) and fluvastatin (FLU, 33.8% ± 0.6%; P < 0.01). Moreover, the combination of EVR and FLU showed a greater suppressive effect on HLA‐DR expression. In contrast, cyclosporine, tacrolimus, mycophenolic acid, and prednisolone did not exhibit any significant suppressive effect. FLU, but not EVR, suppressed mRNA of HLA‐DR. Imaging analysis revealed that HLA‐DR expressed in cytosol or on the cell surface was repressed by EVR (cytosol: 58.6% ± 4.9%, P < 0.01; cell surface: 80.9% ± 4.0%, P < 0.01) and FLU (cytosol: 19.0% ± 3.4%, P < 0.01; cell surface: 48.3% ± 4.8%, P < 0.01). These data indicated that FLU and EVR suppressed IFN‐γ‐induced HLA‐DR expression at the transcriptional and post‐translational level, respectively, suggesting a potential approach for alleviating DSA‐related issues in organ transplantation.
Collapse
Affiliation(s)
- Akihiro Maenaka
- Department of Pharmacy, Aichi Medical University School of Medicine, Nagakute, Japan.,Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Iwasaki Kenta
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yuko Miwa
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Wataru Ohashi
- Division of Biostatistics, Clinical Research Center, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kosei Horimi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yutaka Matsuoka
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Masafumi Ohnishi
- Department of Pharmacy, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kazuharu Uchida
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
39
|
Tomita Y, Iwadoh K, Ogawa Y, Miki K, Kato Y, Kai K, Sannomiya A, Koyama I, Kitajima K, Nakajima I, Fuchinoue S. Single fixed low-dose rituximab as induction therapy suppresses de novo donor-specific anti-HLA antibody production in ABO compatible living kidney transplant recipients. PLoS One 2019; 14:e0224203. [PMID: 31644555 PMCID: PMC6808551 DOI: 10.1371/journal.pone.0224203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 10/08/2019] [Indexed: 01/09/2023] Open
Abstract
This study was conducted to evaluate de novo donor-specific anti-human leukocyte antigen (HLA) antibody (dnDSA) production leading to antibody-mediated rejection (ABMR) after rituximab induction in non-sensitized ABO-compatible living kidney transplantation (ABO-CLKTx). During 2008-2015, 318 ABO-CLKTx were performed at the Department of Surgery III at Tokyo Women's Medical University Hospital. To reduce confounding factors, we adopted a propensity score analysis, which was applied with adjustment for age, gender, duration of pretransplant dialysis, HLA mismatch count, preformed DSA, non-insulin-dependent diabetes mellitus, immunosuppressive treatment, and estimated glomerular filtration rate (eGFR) on postoperative day 7. Using a propensity score matching model (1:1, 115 pairs), we analyzed the long-term outcomes of 230 ABO-CLKTx recipients retrospectively. Recipients were classified into a rituximab-treated (RTX-KTx, N = 115) group and a control group not treated with rituximab (C-KTx, N = 115). During five years, adverse events, survival rates for grafts and patients, and incidence of biopsy-proven acute rejection (BPAR) and dnDSA production for the two groups were monitored and compared. All recipients in the RTX-KTx group received rituximab induction on preoperative day 4 at a single fixed low dose of 100 mg; the CD19+ B cells were eliminated completely before surgery. Of those recipients, 13 (11.3%) developed BPAR; 1 (0.8%) experienced graft loss. By contrast, of C-KTx group recipients, 25 (21.7%) developed BPAR; 3 (2.6%) experienced graft loss. The RTX-KTx group exhibited a significantly lower incidence of BPAR (P = .041) and dnDSA production (13.9% in the RTX-KTx group vs. 26.9% in the C-RTx group, P = .005). Furthermore, lower incidence of CMV infection was detected in the RTX-KTx group than in the C-KTx group (13.9% in the RTX-KTx group vs. 27.0% in the C-KTx group, P = .014). No significant difference was found between groups for several other factors: renal function (P = .384), graft and patient survival (P = .458 and P = .119, respectively), and the respective incidences of BK virus infection (P = .722) and leukopenia (P = .207). During five-year follow-up, single fixed low-dose rituximab therapy is sufficient for ensuring safety, reducing rejection, and suppressing dnDSA production for immunological low-risk non-sensitized ABO-CLKTx.
Collapse
Affiliation(s)
- Yusuke Tomita
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
- * E-mail:
| | - Kazuhiro Iwadoh
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yuichi Ogawa
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsuyuki Miki
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yojiro Kato
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kotaro Kai
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Akihito Sannomiya
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ichiro Koyama
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kumiko Kitajima
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ichiro Nakajima
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | - Shohei Fuchinoue
- Department of Surgery, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
40
|
Tafulo S, Malheiro J, Santos S, Dias L, Almeida M, Martins LS, Pedroso S, Mendes C, Lobato L, Castro-Henriques A. Degree of HLA class II eplet mismatch load improves prediction of antibody-mediated rejection in living donor kidney transplantation. Hum Immunol 2019; 80:966-975. [PMID: 31604581 DOI: 10.1016/j.humimm.2019.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/05/2019] [Accepted: 09/27/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND HLA mismatching is a well known risk factor for worst outcomes in kidney transplantation. METHODS In the present study, HLA antigen and eplet mismatches were determined in 151 living donor-recipient pairs transplanted between 2007 and 2014 and rejection episodes and graft survival were evaluated. RESULTS We found that high HLA-II eplet mismatch load (EpMM ≥ 13, versus low EpMM ≤ 5), was an independent predictor of AMR (adjusted HR = 14.839; P = 0.011), while HLA-II AgMM was not. We also showed that HLA-II EpMM load was a significant better predictor of AMR than AgMM (c-statistic = 0.064; P = 0.023). After discriminating HLA-II into HLA-DR and HLA-DQ loci we demonstrated that high versus low eplet mismatch load for HLA-DR (T3 ≥ 6 versus T = 0-1, p = 0.013) and HLA-DQ (T3 ≥ 7 versus T = 0-1, p = 0.009) are independent predictors for AMR. HLA-II EpMM increased discrimination performance of the classical HLA-II AgMM risk model (IDI, 0.061, 95%CI: 0.005-0.195) for AMR. Compared with AgMM, HLA-II eplet model adequately reclassified 13 of 17 patients (76.5%) with AMR and 92 of 134 patients (68.7%) without AMR (cfNRI, 0.785, 95%CI: 0.300-1.426). CONCLUSIONS Our study evidences that eplet-based matching is a refinement of the classical HLA antigen mismatch analysis in LDKT and is a potential biomarker for personalized assessment of alloimmune risk.
Collapse
Affiliation(s)
- Sandra Tafulo
- Blood and Transplantation Center of Porto, Portuguese Institute for Blood and Transplantation, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.
| | - Jorge Malheiro
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - Sofia Santos
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - Leonídio Dias
- Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - Manuela Almeida
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - La Salete Martins
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - Sofia Pedroso
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - Cecília Mendes
- Blood and Transplantation Center of Porto, Portuguese Institute for Blood and Transplantation, Porto, Portugal
| | - Luísa Lobato
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal; Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| | - António Castro-Henriques
- Department of Nephrology, Hospital de Santo António, Centro Hospitalar Universitário do Porto, Portugal
| |
Collapse
|
41
|
Iwasaki K, Kitahata N, Hiramitsu T, Yamamoto T, Noda T, Okada M, Narumi S, Watarai Y, Miwa Y, Uchida K, Matsuoka Y, Horimi K, Kobayashi T. Increased CD40L+PD-1+ follicular helper T cells (Tfh) as a biomarker for predicting calcineurin inhibitor sensitivity against Tfh-mediated B-cell activation/antibody production after kidney transplantation. Int Immunol 2019; 30:345-355. [PMID: 29878122 DOI: 10.1093/intimm/dxy039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/30/2018] [Indexed: 11/13/2022] Open
Abstract
It is unclear to what extent the development of follicular helper T cells (Tfh) and de novo donor-specific human leukocyte antigen antibody (DSA) production could be influenced by immunosuppressive agents, particularly calcineurin inhibitor (CNI; cyclosporine or tacrolimus), after kidney transplantation. Here, the effects of immunosuppressive agents on Tfh-mediated B-cell activation and antibody production were investigated. In vitro circulating Tfh (cTfh; memory CD4+CXCR5+)/B-cell (CD19+) co-culture assays revealed that CNI considerably inhibited cTfh-mediated B-cell activation and IgG antibody secretion through the suppression of IL-21 and IL-2. Both IL-21 and CD40L up-regulated IL-2 receptors (CD25) on B cells, and anti-CD25 antibody induced apoptosis of activated B cells, resulting in the inhibition of IgG production. The frequency of cTfh-expressed CD40L and PD-1 was elevated in patients with de novo DSA 1 year after transplantation. The degree of inhibition by CNI was dependent on Staphylococcal enterotoxin B-induced CD40L+PD-1+ cTfh up-regulation level. Our data demonstrate that CD40L+PD-1+cTfh could be a marker to implicate individual difference in CNI sensitivity for Tfh-mediated B-cell activation in kidney transplantation.
Collapse
Affiliation(s)
- Kenta Iwasaki
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Nana Kitahata
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takahisa Hiramitsu
- Nagoya Daini Red Cross Hospital, Department of Nephrology, 2-9 Myoken-cho, Showa-ku, Nagoya, Japan
| | - Takayuki Yamamoto
- Nagoya Daini Red Cross Hospital, Department of Nephrology, 2-9 Myoken-cho, Showa-ku, Nagoya, Japan
| | - Takayuki Noda
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Manabu Okada
- Nagoya Daini Red Cross Hospital, Department of Nephrology, 2-9 Myoken-cho, Showa-ku, Nagoya, Japan
| | - Shunji Narumi
- Nagoya Daini Red Cross Hospital, Department of Nephrology, 2-9 Myoken-cho, Showa-ku, Nagoya, Japan
| | - Yoshihiko Watarai
- Nagoya Daini Red Cross Hospital, Department of Nephrology, 2-9 Myoken-cho, Showa-ku, Nagoya, Japan
| | - Yuko Miwa
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Kazuharu Uchida
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yutaka Matsuoka
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Kosei Horimi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| |
Collapse
|
42
|
Abstract
Background Antibody-mediated rejection is a major cause of premature graft loss in kidney transplantation. Multiple scoring systems are available to assess the HLA mismatch between donors and recipients at the molecular level; however, their correlation with the development of de novo donor-specific antibody (dnDSA) has not been compared in recipients on active immunosuppression. Methods HLA-DRβ1/3/4/5/DQα1β1 molecular mismatch was determined using eplet analysis, amino acid mismatch, and electrostatic mismatch for 596 renal transplant recipients and correlated with HLA-DR/DQ dnDSA development. The molecular mismatch scores were evaluated in multivariate models of posttransplant dnDSA-free survival. Results Eplet mismatch correlated with amino acid mismatch and electrostatic mismatch (R2 = 0.85-0.96). HLA-DR dnDSA-free survival correlated with HLA-DR eplet mismatch (hazards ratio [HR], 2.50 per 10 eplets mismatched; P < 0.0001), amino acid mismatch (HR, 1.49 per 10 amino acids mismatched; P < 0.0001), and electrostatic mismatch (HR, 1.23 per 10 units mismatched; P < 0.0001). HLA-DQ dnDSA-free survival correlated with HLA-DQ eplet mismatch (HR, 1.98 per 10 eplets mismatched; P < 0.0001), amino acid mismatch (HR, 1.24 per 10 amino acids mismatched; P < 0.0001), and electrostatic mismatch (HR, 1.14 per 10 units mismatched; P < 0.0001). All 3 methods were significant multivariate correlates of dnDSA development after adjustment for recipient age, baseline immunosuppression, and nonadherence. Conclusions HLA molecular mismatch represents a precise method of alloimmune risk assessment for renal transplant patients. The method used to determine the molecular mismatch is likely to be driven by familiarity and ease of use as highly correlated results are produced by each method. In order to assess alloimmune risk in kidney transplant recipiennts, the authors compare 3 methods evaluating HLA molecular mismatch (HLA-DG eplet, amino acid and electrostatic) which all correlate with adjusted dnDSA development. Supplemental digital content is available in the text.
Collapse
|
43
|
Snanoudj R, Kamar N, Cassuto E, Caillard S, Metzger M, Merville P, Thierry A, Jollet I, Grimbert P, Anglicheau D, Hazzan M, Choukroun G, Hurault De Ligny B, Janbon B, Vuiblet V, Devys A, Le Meur Y, Delahousse M, Morelon E, Bailly E, Girerd S, Amokrane K, Legendre C, Hertig A, Rondeau E, Taupin JL. Epitope load identifies kidney transplant recipients at risk of allosensitization following minimization of immunosuppression. Kidney Int 2019; 95:1471-1485. [PMID: 30955869 DOI: 10.1016/j.kint.2018.12.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 12/07/2018] [Accepted: 12/28/2018] [Indexed: 12/01/2022]
Abstract
Human leukocyte antigen (HLA) mismatching and minimization of immunosuppression are two major risk factors for the development of de novo donor-specific antibodies, which are associated with reduced kidney graft survival. Antibodies do not recognize whole HLA antigens but rather individual epitopes, which are short sequences of amino acids in accessible positions. However, compatibility is still assessed by the simple count of mismatched HLA antigens. We hypothesized that the number of mismatched epitopes, or ("epitope load") would identify patients at the highest risk of developing donor specific antibodies following minimization of immunosuppression. We determined epitope load in 89 clinical trial participants who converted from cyclosporine to everolimus 3 months after kidney transplantation. Twenty-nine participants (32.6%) developed de novo donor specific antibodies. Compared to the number of HLA mismatches, epitope load was more strongly associated with the development of donor specific antibodies. Participants with an epitope load greater than 27 had a 12-fold relative risk of developing donor-specific antibodies compared to those with an epitope load below that threshold. Using that threshold, epitope load would have missed only one participant who subsequently developed donor specific antibodies, compared to 8 missed cases based on a 6-antigen mismatch. DQ7 was the most frequent antigenic target of donor specific antibodies in our population, and some DQ7 epitopes appeared to be more frequently involved than others. Assessing epitope load before minimizing immunosuppression may be a more efficient tool to identify patients at the highest risk of allosensitization.
Collapse
Affiliation(s)
- Renaud Snanoudj
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Hôpital Foch, Suresnes, France; CESP, INSERM, Université Paris-Sud, UVSQ, Université Paris-Saclay, Villejuif, France.
| | - Nassim Kamar
- Service de Néphrologie et Transplantation d'organe, CHU Rangueil, Toulouse, France
| | | | - Sophie Caillard
- Service de Néphrologie et Transplantation, CHU de Strasbourg, Strasbourg, France
| | - Marie Metzger
- CESP, INSERM, Université Paris-Sud, UVSQ, Université Paris-Saclay, Villejuif, France
| | - Pierre Merville
- Service de Néphrologie et Transplantation, Hôpital Pellegrin, Bordeaux, France
| | - Antoine Thierry
- Service de Néphrologie et Transplantation, CHU de Poitiers, Poitiers, France
| | | | - Philippe Grimbert
- Service de Néphrologie et Transplantation, Hôpital Henri Mondor, Créteil, France
| | - Dany Anglicheau
- Service de Transplantation et Néphrologie, Hôpital Necker, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Marc Hazzan
- Service de Néphrologie, Hôpital Claude Huriez, CHRU de Lille, Lille, France
| | | | | | - Bénedicte Janbon
- Service de Néphrologie et Transplantation, CHU de Grenoble, Grenoble, France
| | - Vincent Vuiblet
- Service de Néphrologie, Hôpital Maison Blanche, Reims, France
| | - Anne Devys
- Etablissement français du sang - Centre Pays de Loire, Angers, France
| | - Yann Le Meur
- Service de Néphrologie, CHU de Brest, Brest, France
| | - Michel Delahousse
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Hôpital Foch, Suresnes, France
| | - Emmanuel Morelon
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Elodie Bailly
- Service de Néphrologie, Hôpital Bretonneau, CHU de Tours, Tours, France
| | - Sophie Girerd
- Service de Néphrologie, CHU Nancy Brabois, Nancy, France
| | - Kahina Amokrane
- Laboratoire d'Immunologie et Histocompatibilité Hôpital Saint-Louis, Paris, France; INSERM UMRs 1160, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Christophe Legendre
- Service de Transplantation et Néphrologie, Hôpital Necker, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandre Hertig
- Service de Néphrologie et Transplantation, Hôpital Tenon, Paris, France
| | - Eric Rondeau
- Service de Néphrologie et Transplantation, Hôpital Tenon, Paris, France
| | - Jean-Luc Taupin
- Laboratoire d'Immunologie et Histocompatibilité Hôpital Saint-Louis, Paris, France; INSERM UMRs 1160, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| |
Collapse
|
44
|
Padet L, Dieudé M, Karakeussian‐Rimbaud A, Yang B, Turgeon J, Cailhier J, Cardinal H, Hébert M. New insights into immune mechanisms of antiperlecan/LG3 antibody production: Importance of T cells and innate B1 cells. Am J Transplant 2019; 19:699-712. [PMID: 30129231 PMCID: PMC6519043 DOI: 10.1111/ajt.15082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/25/2018] [Accepted: 07/29/2018] [Indexed: 01/25/2023]
Abstract
Autoantibodies against perlecan/LG3 (anti-LG3) have been associated with increased risks of delayed graft function, acute rejection, and reduced long-term survival. High titers of anti-LG3 antibodies have been found in de novo renal transplants recipients in the absence of allosensitizing or autoimmune conditions. Here, we seek to understand the pathways controlling anti-LG3 production prior to transplantation. Mice immunized with recombinant LG3 produce concomitantly IgM and IgG anti-LG3 antibodies suggesting a memory response. ELISpot confirmed the presence of LG3-specific memory B cells in nonimmunized mice. Purification of B1 and B2 subtypes identified peritoneal B1 cells as the major source of memory B cells reactive to LG3. Although nonimmunized CD4-deficient mice were found to express LG3-specific memory B cells, depletion of CD4+ T cells in wild type mice during immunization significantly decreased anti-LG3 production. These results demonstrate that B cell memory to LG3 is T cell independent but that production of anti-LG3 antibodies requires T cell help. Further supporting an important role for T cells in controlling anti-LG3 levels, we found that human renal transplant recipients show a significant decrease in anti-LG3 titers upon the initiation of CNI-based immunosuppression. Collectively, these results identify T cell targeting interventions as a means of reducing anti-LG3 levels in renal transplant patients.
Collapse
Affiliation(s)
- Lauriane Padet
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada,Université de MontréalMontrealQuebecCanada
| | - Mélanie Dieudé
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Annie Karakeussian‐Rimbaud
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Bing Yang
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada,Université de MontréalMontrealQuebecCanada
| | - Julie Turgeon
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Jean‐François Cailhier
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Héloïse Cardinal
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Marie‐Josée Hébert
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada,Université de MontréalMontrealQuebecCanada
| |
Collapse
|
45
|
Okada M, Watarai Y, Iwasaki K, Futamura K, Yamamoto T, Hiramitsu T, Tsujita M, Goto N, Narumi S, Takeda A, Kobayashi T. Lower incidence of de novo donor-specific antibodies against HLA-DR in ABO-incompatible renal transplantation. Hum Immunol 2018; 80:169-175. [PMID: 30552908 DOI: 10.1016/j.humimm.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/25/2018] [Accepted: 12/09/2018] [Indexed: 12/12/2022]
Abstract
Recently, in vitro experiments have demonstrated that anti-blood group A/B antibody binding to endothelial cells induce a protective effect against antibody-mediated injury. This study aimed to clarify the potential clinical benefit of ABO incompatibility in donor-specific HLA antibody (DSA)-induced chronic antibody-mediated rejection (ABMR). We enrolled 215 ABO-incompatible renal transplant (ABO-I) and 467 ABO-identical/compatible renal transplant recipients (ABO-Id/C). The prevalence of de novo DSA production and incidence of biopsy-proven chronic ABMR were compared between the two groups. The incidence of DR-associated de novo DSA was significantly lower in ABO-I than in ABO-Id/C (P = 0.028). Diagnostic biopsy for ABMR was conducted in 54 patients (11 ABO-I and 43 ABO-Id/C). Biopsy-proven chronic ABMR was lower in ABO-I than in ABO-Id/C (27.3% [3/11] vs. 44.2% [19/43]) patients. Our findings suggest that ABO incompatibility may cause low production of DR-associated de novo DSA, possibly resulting in a reduced incidence of chronic ABMR.
Collapse
Affiliation(s)
- Manabu Okada
- Department of Transplant Surgery, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan; Department of Renal Transplant Surgery, Aichi Medical University, School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Yoshihiko Watarai
- Department of Transplant Surgery, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Kenta Iwasaki
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University, School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Kenta Futamura
- Department of Transplant Internal Medicine, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya, 466-8650, Japan
| | - Takayuki Yamamoto
- Department of Transplant Surgery, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Takahisa Hiramitsu
- Department of Transplant Surgery, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Makoto Tsujita
- Department of Transplant Internal Medicine, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya, 466-8650, Japan
| | - Norihiko Goto
- Department of Transplant Internal Medicine, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya, 466-8650, Japan
| | - Shunji Narumi
- Department of Transplant Surgery, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Asami Takeda
- Department of Nephrology, Kidney Disease Center, Nagoya Daini Red Cross Hospital, 2-9 Myoken-cho, Showa-ku, Nagoya 466-8650, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University, School of Medicine, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan.
| |
Collapse
|
46
|
Early Diagnosis and Treatment of Subclinical AMR Is Vital for Improving Clinical Outcomes. Transplantation 2018; 103:1542-1543. [PMID: 30507743 DOI: 10.1097/tp.0000000000002567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Koefoed-Nielsen P, Møller BK. Donor-specific anti-HLA antibodies by solid phase immunoassays: advantages and technical concerns. Int Rev Immunol 2018; 38:95-105. [DOI: 10.1080/08830185.2018.1525367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Bjarne Kuno Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
48
|
Karahan GE, Claas FHJ, Heidt S. Technical challenges and clinical relevance of single antigen bead C1q/C3d testing and IgG subclass analysis of human leukocyte antigen antibodies. Transpl Int 2018; 31:1189-1197. [DOI: 10.1111/tri.13327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/21/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Gonca E. Karahan
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden the Netherlands
| | - Frans H. J. Claas
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden the Netherlands
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood Transfusion; Leiden University Medical Center; Leiden the Netherlands
| |
Collapse
|
49
|
Jung HY, Kim SH, Seo MY, Cho SY, Yang Y, Choi JY, Cho JH, Park SH, Kim YL, Kim HK, Huh S, Won DI, Kim CD. Characteristics and Clinical Significance of De Novo Donor-Specific Anti-HLA Antibodies after Kidney Transplantation. J Korean Med Sci 2018; 33:e217. [PMID: 30127706 PMCID: PMC6097072 DOI: 10.3346/jkms.2018.33.e217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/16/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The association of de novo donor-specific anti-human leukocyte antigens (HLA) antibodies (DSA) and development of antibody-mediated rejection (AMR) in kidney transplant recipients (KTRs) is still undetermined. METHODS We prospectively screened de novo DSA in 167 KTRs during 32 months after kidney transplantation (KT). Timing of DSA detection was at 3, 6, and 12 months post-transplant and annually thereafter and when clinically indicated. DSA levels were determined by Luminex assays and expressed as mean fluorescence intensity (MFI). We evaluated the incidence, characteristics of DSA, and association between DSA and tacrolimus trough levels or AMR. RESULTS De novo DSA developed in 16 KTRs (9.6%) and acute AMR occurred more commonly in KTRs with de novo DSA compared to KTRs without de novo DSA (18.8% vs. 0%, P < 0.001). All de novo DSA were against class II antigens. The mean number of DSA was 1.8 ± 1.2 and the average MFI of DSA was 7,399 ± 5,470. Tacrolimus trough level during the first 0-2 months after KT was an independent predictor of DSA development (hazard ratio, 0.70; 95% confidence interval, 0.50-0.99; P = 0.043). No differences were found in the number of DSA, average MFI of DSA, and tacrolimus levels during the first year between de novo DSA-positive KTRs with AMR and those without AMR. CONCLUSION The results of our study suggest that monitoring of DSA and maintaining proper tacrolimus levels are essential to prevent AMR during the initial period after KT.
Collapse
Affiliation(s)
- Hee-Yeon Jung
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Su-Hee Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Min-Young Seo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sun-Young Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Youngae Yang
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ji-Young Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jang-Hee Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sun-Hee Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yong-Lim Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hyung-Kee Kim
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Seung Huh
- Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Dong Il Won
- Department of Clinical Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Chan-Duck Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
50
|
Zhao D, Liao T, Li S, Zhang Y, Zheng H, Zhou J, Han F, Dong Y, Sun Q. Mouse Model Established by Early Renal Transplantation After Skin Allograft Sensitization Mimics Clinical Antibody-Mediated Rejection. Front Immunol 2018; 9:1356. [PMID: 30022978 PMCID: PMC6039569 DOI: 10.3389/fimmu.2018.01356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/31/2018] [Indexed: 01/03/2023] Open
Abstract
Antibody-mediated rejection (AMR) is the main barrier to renal graft survival, and mouse renal AMR models are important to study this process. Current mouse models are established by priming the recipient to donor skin for over 7 days before kidney transplantation. The robustness of AMR in these cases is too strong to mimic clinical AMR and it is unclear why altering the priming times ranging from 7 to 91 days fails to reduce the AMR potency in these models. In the present study, we found that the donor-recipient combination and skin graft size were determinants of donor-specific antibody (DSA) development patterns after skin transplantation. DSA-IgG was sustained for over 100 days after skin challenge, accounting for an identical AMR robustness upon different skin priming times over 7 days. However, decreasing the skin priming time within 7 days attenuated the robustness of subsequent renal allograft AMR in C3H to Balb/c mice. Four-day skin priming guaranteed that recipients develop acute renal AMR mixed with a high ratio of graft-infiltrating macrophages, renal grafts survived for a mean of 6.4 ± 2.1 days, characterized by typical AMR histological changes, such as glomerulitis, peritubular capillary (PTC) dilation, and capillaritis, deposition of IgG and C3d in PTCs, but less prevalence of microthrombus, whereas the cellular rejection histological change of tubulitis was absent to mild. With this scheme, we also found that the renal AMR model can be developed using common mouse strains such as C57BL/6 and Balb/c, with mean prolonged renal graft survival times of 14.4 ± 5.0 days. Finally, we proved that donor-matched skin challenge after kidney transplantation did not strongly affect DSA development and kidney graft outcome. These findings may facilitate an understanding and establishment of mouse renal allograft AMR models and promote AMR-associated studies.
Collapse
Affiliation(s)
- Daqiang Zhao
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tao Liao
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Siwen Li
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yannan Zhang
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haofeng Zheng
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Zhou
- Department of Pathology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fei Han
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Dong
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiquan Sun
- Division of Kidney Transplantation, Department of Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|