1
|
Mattoo A, Jaffe IS, Keating B, Montgomery RA, Mangiola M. Improving long-term kidney allograft survival by rethinking HLA compatibility: from molecular matching to non-HLA genes. Front Genet 2024; 15:1442018. [PMID: 39415982 PMCID: PMC11480002 DOI: 10.3389/fgene.2024.1442018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Optimizing immunologic compatibility in organ transplantation extends beyond the conventional approach of Human Leukocyte Antigen (HLA) antigen matching, which exhibits significant limitations. A broader comprehension of the roles of classical and non-classical HLA genes in transplantation is imperative for enhancing long-term graft survival. High-resolution molecular HLA genotyping, despite its inherent challenges, has emerged as the cornerstone for precise patient-donor compatibility assessment. Leveraging understanding of eplet biology and indirect immune activation, eplet mismatch calculators and the PIRCHE-II algorithm surpass traditional methods in predicting allograft rejection. Understanding minor histocompatibility antigens may also present an opportunity to personalize the compatibility process. While the application of molecular matching in deceased donor organ allocation presents multiple technical, logistical, and conceptual barriers, rendering it premature for mainstream use, several other areas of donor-recipient matching and post-transplant management are ready to incorporate molecular matching. Provision of molecular mismatch scores to physicians during potential organ offer evaluations could potentially amplify long-term outcomes. The implementation of molecular matching in living organ donation and kidney paired exchange programs is similarly viable. This article will explore the current understanding of immunologic matching in transplantation and the potential applications of epitope and non-epitope molecular biology and genetics in clinical transplantation.
Collapse
Affiliation(s)
- Aprajita Mattoo
- *Correspondence: Aprajita Mattoo, ; Ian S. Jaffe, ; Massimo Mangiola,
| | - Ian S. Jaffe
- *Correspondence: Aprajita Mattoo, ; Ian S. Jaffe, ; Massimo Mangiola,
| | | | | | - Massimo Mangiola
- NYU Langone Transplant Institute, New York University Langone Health, New York, NY, United States
| |
Collapse
|
2
|
Aisihaer X, Guo H, Liu C. Interchain disulfide engineering enables the efficient production of functional HLA-DQ-Fc fusion proteins. J Biol Chem 2024; 300:107652. [PMID: 39121997 PMCID: PMC11402769 DOI: 10.1016/j.jbc.2024.107652] [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: 05/02/2024] [Revised: 07/13/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
HLA-DQ molecules drive unwanted alloimmune responses after solid-organ transplants and several autoimmune diseases, including type 1 diabetes and celiac disease. Biologics with HLA molecules as part of the design are emerging therapeutic options for these allo- and autoimmune conditions. However, the soluble α and β chains of class II HLA molecules do not dimerize efficiently without their transmembrane domains, which hinders their production. In this study, we examined the feasibility of interchain disulfide engineering by introducing paired cysteines to juxtaposed positions in the α and β chains of HLA-DQ7, encoded by HLA-DQA1∗05:01 and HLA-DQB1∗03:01 respectively. We identified three variant peptide-HLA-DQ7-Fc fusion proteins (DQ7Fc) with increased expression and production yield, namely Y19C-D6C (YCDC), A83C-E5C (ACEC), and A84C-N33C (ACNC). The mutated residues were conserved across all HLA-DQ proteins and had limited solvent exposure. Further characterizations of the YCDC variant showed that the expression of the fusion protein is peptide-dependent; inclusion of a higher-affinity peptide correlated with increased protein expression. However, high-affinity peptide alone was insufficient for stabilizing the DQ7 complex without the engineered disulfide bond. Multiple DQ7Fc variants demonstrated expected binding characteristics with commercial anti-DQ antibodies in two immunoassays and by a cell-based assay. Lastly, DQ7Fc variants demonstrated dose-dependent killing of DQ7-specific B cell hybridomas in a flow cytometric, complement-dependent cytotoxicity assay. These data support inter-chain disulfide engineering as a novel approach to efficiently producing functional HLA-DQ molecules and potentially other class II HLA molecules as candidate therapeutic agents.
Collapse
Affiliation(s)
| | - Hongjie Guo
- Antiger Therapeutics Inc., St Louis, Missouri, USA.
| | - Chang Liu
- Antiger Therapeutics Inc., St Louis, Missouri, USA.
| |
Collapse
|
3
|
Hu Q, Abu-Khader A, Christian D, Yang G, Berka N. Discovery of potential recombinant alleles HLA-DRB3*02:171 and HLA-DRB5*01:140. HLA 2024; 104:e15697. [PMID: 39291436 DOI: 10.1111/tan.15697] [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: 08/08/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024]
Abstract
Two novel alleles that appear to be generated from recombination between exons 2 and 3.
Collapse
Affiliation(s)
- Qian Hu
- Transplant Laboratory South, Alberta Precision Laboratories, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ahmad Abu-Khader
- Transplant Laboratory South, Alberta Precision Laboratories, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Danielle Christian
- Transplant Laboratory South, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Guang Yang
- Transplant Laboratory South, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Noureddine Berka
- Transplant Laboratory South, Alberta Precision Laboratories, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Diebold M, Mayer KA, Hidalgo L, Kozakowski N, Budde K, Böhmig GA. Chronic Rejection After Kidney Transplantation. Transplantation 2024:00007890-990000000-00858. [PMID: 39192468 DOI: 10.1097/tp.0000000000005187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
In kidney transplantation, ongoing alloimmune processes-commonly triggered by HLA incompatibilities-can trigger chronic transplant rejection, affecting the microcirculation and the tubulointerstitium. Continuous inflammation may lead to progressive, irreversible graft injury, culminating in graft dysfunction and accelerated transplant failure. Numerous experimental and translational studies have delineated a complex interplay of different immune mechanisms driving rejection, with antibody-mediated rejection (AMR) being an extensively studied rejection variant. In microvascular inflammation, a hallmark lesion of AMR, natural killer (NK) cells have emerged as pivotal effector cells. Their essential role is supported by immunohistologic evidence, bulk and spatial transcriptomics, and functional genetics. Despite significant research efforts, a substantial unmet need for approved rejection therapies persists, with many trials yielding negative outcomes. However, several promising therapies are currently under investigation, including felzartamab, a monoclonal antibody targeting the surface molecule CD38, which is highly expressed in NK cells and antibody-producing plasma cells. In an exploratory phase 2 trial in late AMR, this compound has demonstrated potential in resolving molecular and morphologic rejection activity and injury, predominantly by targeting NK cell effector function. These findings inspire hope for effective treatments and emphasize the necessity of further pivotal trials focusing on chronic transplant rejection.
Collapse
Affiliation(s)
- Matthias Diebold
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Katharina A Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Luis Hidalgo
- HLA Laboratory, Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | - Klemens Budde
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Wong ETY, Pochinco D, Vathsala A, Koh WK, Lim A, Sran HK, D'Costa MR, Chang ZY, Nickerson PW, Wiebe C. HLA-DR/DQ eplet mismatch predicts de novo donor-specific antibody development in multi-ethnic Southeast Asian kidney transplant recipients on different immunosuppression regimens. Front Genet 2024; 15:1447141. [PMID: 39262421 PMCID: PMC11387181 DOI: 10.3389/fgene.2024.1447141] [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: 06/11/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Eplet mismatch has been recognized as a more precise strategy for determining HLA compatibility by analyzing donor-recipient HLA differences at the molecular level. However, predicting post-transplant alloimmunity using single-molecule eplet mismatch categories has not been validated in Asian cohorts. We examined a cohort of Southeast Asian kidney transplant recipients (n = 234) to evaluate HLA-DR/DQ eplet mismatch as a predictor of de novo donor-specific antibody (dnDSA) development. HLA-DR/DQ single-molecule eplet mismatch was quantified using HLA Matchmaker, and we utilized previously published HLA-DR/DQ eplet mismatch thresholds to categorize recipients into alloimmune risk groups and evaluate their association with dnDSA development. Recognizing that the predominance of cyclosporine use (71%) may alter published eplet mismatch thresholds derived from a largely tacrolimus-based (87%) cohort, we evaluated cohort-specific thresholds for HLA-DR/DQ single-molecule eplet mismatch categories. Recipient ethnicities included Chinese (65%), Malays (17%), Indians (14%), and others (4%). HLA-DR/DQ dnDSA developed in 29/234 (12%) recipients after a median follow-up of 5.4 years, including against isolated HLA-DR (n = 7), isolated HLA-DQ (n = 11), or both (n = 11). HLA-DR/DQ single-molecule eplet mismatch risk categories correlated with dnDSA-free survival (p = 0.001) with low-risk recipients having a dnDSA prevalence of 1% over 5 years. The cohort-specific alloimmune risk categories improved correlation with HLA-DR/DQ dnDSA-free survival and remained significant after adjusting for calcineurin inhibitor and anti-metabolite immunosuppression (p < 0.001). We validated the performance of single-molecule eplet mismatch categories as a prognostic biomarker for HLA-DR/DQ dnDSA development in a cohort of predominantly Asian kidney transplant recipients after adjusting for different immunosuppression regimens.
Collapse
Affiliation(s)
- Emmett Tsz Yeung Wong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
- Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | - Anantharaman Vathsala
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Wee Kun Koh
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Amy Lim
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Hersharan Kaur Sran
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Matthew Ross D'Costa
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Zi Yun Chang
- National University Centre for Organ Transplantation, National University Hospital, Singapore, Singapore
| | - Peter W Nickerson
- Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Shared Health Services Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Shared Health Services Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
6
|
Gramkow AM, Baatrup JH, Gramkow ET, Thiesson HC, Koefoed-Nielsen P. Association of HLA B- and T-cell molecular mismatches with HLA antibodies, rejection, and graft survival in pediatric kidney transplantation. Pediatr Transplant 2024; 28:e14773. [PMID: 38808702 DOI: 10.1111/petr.14773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 04/01/2024] [Accepted: 04/18/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Optimizing graft survival and diminishing human leukocyte antigen (HLA) sensitization are essential for pediatric kidney transplant recipients. More precise HLA matching predicting epitope mismatches could reduce alloreactivity. We investigated the association of predicted HLA B- and T-cell molecular mismatches with the formation of de novo donor-specific antibodies, HLA antibodies, rejection, and graft survival. METHODS Forty-nine pediatric kidney transplant recipients transplanted from 2009 to 2020 were retrospectively studied. Donors and recipients were high-resolution HLA typed, and recipients were screened for HLA antibodies posttransplant. HLA-EMMA (HLA Epitope MisMatch Algorithm) and PIRCHE-II (Predicted Indirectly ReCognizable HLA Epitopes) predicted the molecular mismatches. The association of molecular mismatches and the end-points was explored with logistic regression. RESULTS Five recipients (11%) developed de novo donor-specific antibodies. All five had de novo donor-specific antibodies against HLA class II, with four having HLA-DQ antibodies. We found no associations between PIRCHE-II or HLA-EMMA with de novo donor-specific antibodies, HLA sensitization, graft loss, or rejection. However, we did see a tendency towards an increased odds ratio in PIRCHE-II predicting de novo donor-specific antibodies formation, with an odds ratio of 1.12 (95% CI: 0.99; 1.28) on HLA class II. CONCLUSION While the study revealed no significant associations between the number of molecular mismatches and outcomes, a notable trend was observed - indicating a reduced risk of dnDSA formation with improved molecular match. It is important to acknowledge, however, that the modest population size and limited observed outcomes preclude us from making definitive conclusions.
Collapse
Affiliation(s)
- Ann-Maria Gramkow
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Johanne H Baatrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Emilie T Gramkow
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine - Cancer and Inflammation, University of Southern Denmark, Odense, Denmark
| | - Helle C Thiesson
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
7
|
Jucaud V. Allogeneic HLA Humoral Immunogenicity and the Prediction of Donor-Specific HLA Antibody Development. Antibodies (Basel) 2024; 13:61. [PMID: 39189232 PMCID: PMC11348167 DOI: 10.3390/antib13030061] [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: 04/08/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
The development of de novo donor-specific HLA antibodies (dnDSAs) following solid organ transplantation is considered a major risk factor for poor long-term allograft outcomes. The prediction of dnDSA development is a boon to transplant recipients, yet the assessment of allo-HLA immunogenicity remains imprecise. Despite the recent technological advances, a comprehensive evaluation of allo-HLA immunogenicity, which includes both B and T cell allorecognition, is still warranted. Recent studies have proposed using mismatched HLA epitopes (antibody and T cell) as a prognostic biomarker for humoral alloimmunity. However, the identification of immunogenic HLA mismatches has not progressed despite significant improvements in the identification of permissible mismatches. Certainly, the prediction of dnDSA development may benefit permissible HLA mismatched organ transplantations, personalized immunosuppression, and clinical trial design. However, characteristics that go beyond the listing of mismatched HLA antibody epitopes and T cell epitopes, such as the generation of HLA T cell epitope repertoires, recipient's HLA class II phenotype, and immunosuppressive regiments, are required for the precise assessment of allo-HLA immunogenicity.
Collapse
Affiliation(s)
- Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367, USA
| |
Collapse
|
8
|
Liefeldt L, Waiser J, Bachmann F, Budde K, Friedersdorff F, Halleck F, Lachmann N, Peters R, Rudolph B, Ünlü S, Wu K, Glander P. Long-Term Outcome after Early Mammalian Target of Rapamycin Inhibitor-Based Immunosuppression in Kidney Transplant Recipients. J Clin Med 2024; 13:4305. [PMID: 39124572 PMCID: PMC11313631 DOI: 10.3390/jcm13154305] [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: 06/17/2024] [Revised: 07/13/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Background: The use of mammalian target of rapamycin inhibitors (mTORis) in kidney transplantation increases the risk of donor-specific human leukocyte antigen (HLA) antibody formation and rejection. Here, we investigated the long-term consequences of early mTORi treatment compared to calcineurin inhibitor (CNI) treatment. Methods: In this retrospective single-center analysis, key outcome parameters were compared between patients participating in randomized controlled immunosuppression trials between 1998 and 2011, with complete follow-up until 2018. The outcomes of eligible patients on a CNI-based regimen (n = 384) were compared with those of patients randomized to a CNI-free mTORi-based regimen (n = 81) and 76 patients randomized to a combination of CNI and mTORi treatments. All data were analyzed according to the intention-to-treat (ITT) principle. Results: Deviation from randomized immunosuppression for clinical reasons occurred significantly more often and much earlier in both mTORi-containing regimens than in the CNI treatment. Overall patient survival, graft survival, and death-censored graft survival did not differ between the treatment groups. Donor-specific HLA antibody formation and BPARs were significantly more common in both mTORi regimens than in the CNI-based immunosuppression. Conclusions: The tolerability and efficacy of the mTORi treatment in kidney graft recipients are inferior to those of CNI-based immunosuppression, while the long-term patient and graft survival rates were similar.
Collapse
Affiliation(s)
- Lutz Liefeldt
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| | - Johannes Waiser
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| | - Friederike Bachmann
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| | - Klemens Budde
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| | - Frank Friedersdorff
- Department of Urology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (F.F.); (R.P.)
| | - Fabian Halleck
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| | - Nils Lachmann
- Centre for Tumor Medicine, H&I Laboratory, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (N.L.); (S.Ü.)
| | - Robert Peters
- Department of Urology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany; (F.F.); (R.P.)
| | - Birgit Rudolph
- Department of Pathology, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Sinem Ünlü
- Centre for Tumor Medicine, H&I Laboratory, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (N.L.); (S.Ü.)
| | - Kaiyin Wu
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| | - Petra Glander
- Department of Nephrology and Internal Intensive Care Medicine, Charité—Universitätsmedizin Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany; (J.W.); (F.B.); (K.B.); (F.H.); (K.W.); (P.G.)
| |
Collapse
|
9
|
Cohen GS, Gareau AJ, Kallarakal MA, Farooq T, Bettinotti MP, Sullivan HC, Madbouly A, Krummey SM. HLA Genotype Imputation Results in Largely Accurate Epitope Mismatch Risk Categorization Across Racial Groups. Transplant Direct 2024; 10:e1639. [PMID: 38911277 PMCID: PMC11191912 DOI: 10.1097/txd.0000000000001639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/09/2024] [Indexed: 06/25/2024] Open
Abstract
Background Biomarkers that predict posttransplant alloimmunity could lead to improved long-term graft survival. Evaluation of the number of mismatched epitopes between donor and recipient HLA proteins, termed molecular mismatch analysis, has emerged as an approach to classify transplant recipients as having high, intermediate, or low risk of graft rejection. When high-resolution genotypes are unavailable, molecular mismatch analysis requires algorithmic assignment, or imputation, of a high-resolution genotyping. Although imputation introduces inaccuracies in molecular mismatch analyses, it is unclear whether these inaccuracies would impact the clinical risk assessment for graft rejection. Methods Using renal transplant patients and donors from our center, we constructed cohorts of surrogate donor-recipient pairs with high-resolution and low-resolution HLA genotyping that were racially concordant or discordant. We systemically assessed the impact of imputation on molecular mismatch analysis for cohorts of 180-200 donor-recipient pairs for each of 4 major racial groups. We also evaluated the effect of imputation for a racially diverse validation cohort of 35 real-world renal transplant pairs. Results In the surrogate donor-recipient cohorts, imputation preserved the molecular mismatch risk category for 90.5%-99.6% of racially concordant donor-recipient pairs and 92.5%-100% of racially discordant pairs. In the validation cohort, which comprised 72% racially discordant pairs, we found that imputation preserved the molecular mismatch risk category for 97.1% of pairs. Conclusions Overall, these data demonstrate that imputation preserves the molecular mismatch risk assessment in the vast majority of cases and provides evidence supporting imputation in the performance of molecular mismatch analysis for clinical assessment.
Collapse
Affiliation(s)
- Gregory S. Cohen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alison J. Gareau
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Johns Hopkins Immunogenetics Laboratory, Baltimore, MD
| | | | - Tayyiaba Farooq
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Maria P. Bettinotti
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Johns Hopkins Immunogenetics Laboratory, Baltimore, MD
| | - H. Cliff Sullivan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Abeer Madbouly
- National Marrow Donor Program/Be The Match, Minneapolis, MN
- Center for International Blood and Marrow Transplant Research, Minneapolis, MN
| | - Scott M. Krummey
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Johns Hopkins Immunogenetics Laboratory, Baltimore, MD
| |
Collapse
|
10
|
Santos E, Spensley K, Gunby N, Worthington J, Roufosse C, Anand A, Willicombe M. Application of HLA molecular level mismatching in ethnically diverse kidney transplant recipients receiving a steroid-sparing immunosuppression protocol. Am J Transplant 2024; 24:1218-1232. [PMID: 38403189 DOI: 10.1016/j.ajt.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/27/2024]
Abstract
Defining HLA mismatch at the molecular compared with the antigen level has been shown to be superior in predicting alloimmune responses, although data from across different patient populations are lacking. Using HLA-Matchmaker, HLA-EMMA and PIRCHE-II, this study reports on the association between molecular mismatch (MolMM) and de novo donor-specific antibody (dnDSA) in an ethnically diverse kidney transplant population receiving a steroid-sparing immunosuppression protocol. Of the 419 patients, 51 (12.2%) patients had dnDSA. De novo DSA were seen more frequently with males, primary transplants, patients receiving tacrolimus monotherapy, and unfavorably HLA-matched transplants. There was a strong correlation between MolMM load and antigen mismatch, although significant variation of MolMM load existed at each antigen mismatch. MolMM loads differed significantly by recipient ethnicity, although ethnicity alone was not associated with dnDSA. On multivariate analysis, increasing MolMM loads associated with dnDSA, whereas antigen mismatch did not. De novo DSA against 8 specific epitopes occurred at high frequency; of the 51 patients, 47 (92.1%) patients with dnDSA underwent a pretreatment biopsy, with 21 (44.7%) having evidence of alloimmune injury. MolMM has higher specificity than antigen mismatching at identifying recipients who are at low risk of dnDSA while receiving minimalist immunosuppression. Immunogenicity consideration is important, with more work needed on identification, especially across different ethnic groups.
Collapse
Affiliation(s)
- Eva Santos
- Histocompatibility and Immunogenetics Laboratory, Northwest London Pathology NHS Trust, Hammersmith Hospital, London, UK
| | - Katrina Spensley
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK; Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Nicola Gunby
- Histocompatibility and Immunogenetics Laboratory, Northwest London Pathology NHS Trust, Hammersmith Hospital, London, UK
| | - Judith Worthington
- Transplantation Laboratory, Manchester Royal Infirmary, Oxford Road, Manchester, UK
| | - Candice Roufosse
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK; Department of Histopathology, Northwest London Pathology NHS Trust, Charing Cross Hospital, London, UK
| | - Arthi Anand
- Histocompatibility and Immunogenetics Laboratory, Northwest London Pathology NHS Trust, Hammersmith Hospital, London, UK
| | - Michelle Willicombe
- Imperial College Renal and Transplant Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK; Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK.
| |
Collapse
|
11
|
Mankowski MA, Gragert L, Segev DL, Montgomery R, Gentry SE, Mangiola M. Balancing Equity and HLA Matching in Deceased-Donor Kidney Allocation with Eplet Mismatch. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.13.23290644. [PMID: 38947023 PMCID: PMC11213093 DOI: 10.1101/2024.06.13.23290644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Prioritization of HLA antigen-level matching in the US kidney allocation system intends to improve post-transplant survival but causes racial disparities and thus has been substantially de-emphasized. Recently, molecular matching based on eplets has been found to improve risk stratification compared to antigen matching. Methods To assign eplets unambiguously, we utilized a cohort of 5193 individuals with high resolution allele-level HLA genotypes from the National Kidney Registry. Using repeated random sampling to simulate donor-recipient genotype pairings based on the ethnic composition of the historical US deceased donor pool, we profiled the percentage of well-matched donors for candidates by ethnicity. Results The percentage of well-matched donors with zero-DR/DQ eplet mismatch was 3-fold less racially disparate for Black and Asian candidates than percentage of donors with zero-ABDR antigen mismatches, and 2-fold less racially disparate for Latino candidates. For other HLA antigen and eplet mismatch thresholds, the percentage of well-matched donors was more similar across candidate ethnic groups. Conclusions Compared to the current zero-ABDR antigen mismatch, prioritizing a zero-DR/DQ eplet mismatch in allocation would decrease racial disparities and increase the percentage of well-matched donors. High resolution HLA deceased donor genotyping would enable unambiguous assignment of eplets to operationalize molecular mismatch metrics in allocation. Key Points Question: What is the impact of prioritizing low molecular mismatch transplants on racial and ethnic disparities in US deceased-donor kidney allocation, compared to the current prioritization of antigen-level matching?Findings: The lowest-risk eplet mismatch approach decreases racial disparities up to 3-fold compared to lowest-risk antigen mismatch and identifies a larger number of the lowest allo-immune risk donors.Meaning: Prioritizing eplet matching in kidney transplant allocation could both improve outcomes and reduce racial disparities compared to the current antigen matching.
Collapse
|
12
|
Doxiadis I, Loeffler-Wirth H, Lachmann N, Lehmann C. A Short History of B-Cell HLA Epitopes. Transfus Med Hemother 2024; 51:152-157. [PMID: 38867808 PMCID: PMC11166405 DOI: 10.1159/000538447] [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] [Accepted: 03/19/2024] [Indexed: 06/14/2024] Open
Abstract
Background HLA epitopes are currently in the focus of transplantation immunogenetics. The main reason is the complexity of the HLA system with >38,000 alleles, the number of which increases steadily. These alleles are determined by the current state-of-the art typing methods like second- and third-generation sequencing. Screening for HLA antibodies is hampered by the lack of specific target beads with all possible alleles described. Summary A way to circumvent the problem is to define HLA epitopes. The number of antibody-confirmed epitopes, on the other hand, was found to be 72 for HLA class I and 74 for HLA class II. Here, we elaborate on the current knowledge on these HLA epitopes. Absolute definitions of these structures are not yet available. Key Messages Making use of eplets is a comparable way allowing statistical analyses. However, one should keep in mind that the results obtained are approximative or perhaps better associative. Continuous collaboration is needed for the full understanding of the HLA epitopes. The reactivity toward epitopes remains patient-specific.
Collapse
Affiliation(s)
- Ilias Doxiadis
- Laboratory for Transplantation Immunology, University Hospital Leipzig, Faculty of Medicine Leipzig, Leipzig, Germany
| | - Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics, IZBI, Leipzig University, Leipzig, Germany
| | - Nils Lachmann
- Institute for Transfusion Medicine, H and I Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Lehmann
- Laboratory for Transplantation Immunology, University Hospital Leipzig, Faculty of Medicine Leipzig, Leipzig, Germany
| |
Collapse
|
13
|
Ladowski JM, Tector M, Martens G, Wang ZY, Burlak C, Reyes L, Estrada J, Adams A, Tector AJ. Late graft failure of pig-to-rhesus renal xenografts has features of glomerulopathy and recipients have anti-swine leukocyte antigen class I and class II antibodies. Xenotransplantation 2024; 31:e12862. [PMID: 38761019 PMCID: PMC11104517 DOI: 10.1111/xen.12862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 05/20/2024]
Abstract
Prolonged survival in preclinical renal xenotransplantation demonstrates that early antibody mediated rejection (AMR) can be overcome. It is now critical to evaluate and understand the pathobiology of late graft failure and devise new means to improve post xenograft outcomes. In renal allotransplantation the most common cause of late renal graft failure is transplant glomerulopathy-largely due to anti-donor MHC antibodies, particularly anti-HLA DQ antibodies. We evaluated the pig renal xenograft pathology of four long-surviving (>300 days) rhesus monkeys. We also evaluated the terminal serum for the presence of anti-SLA class I and specifically anti-SLA DQ antibodies. All four recipients had transplant glomerulopathy and expressed anti-SLA DQ antibodies. In one recipient tested for anti-SLA I antibodies, the recipient had antibodies specifically reacting with two of three SLA I alleles tested. These results suggest that similar to allotransplantation, anti-MHC antibodies, particularly anti-SLA DQ, may be a barrier to improved long-term xenograft outcomes.
Collapse
Affiliation(s)
| | | | - Gregory Martens
- Department of Surgery, Washington University School of Medicine, St. Louis MO
| | - Zheng Yu Wang
- Department of Surgery, University of Miami School of Medicine, Miami, FL
| | - Chris Burlak
- Department of Surgery, University of Miami School of Medicine, Miami, FL
| | - Luz Reyes
- Department of Surgery, University of Miami School of Medicine, Miami, FL
| | - Jose Estrada
- Department of Surgery, University of Miami School of Medicine, Miami, FL
| | - Andrew Adams
- Department of Surgery, University of Minnesota School of Medicine, Minneapolis, MN
| | - A. Joseph Tector
- Department of Surgery, University of Miami School of Medicine, Miami, FL
| |
Collapse
|
14
|
Jabbour R, Heinzel A, Reindl-Schwaighofer R, Gregorich MG, Regele H, Kozakowski N, Kläger J, Fischer G, Kainz A, Becker JU, Wiebe C, Oberbauer R. Early progression of chronic histologic lesions in kidney transplant biopsies is not associated with HLA histocompatibility. Nephrol Dial Transplant 2024; 39:808-817. [PMID: 37960919 PMCID: PMC11181859 DOI: 10.1093/ndt/gfad246] [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: 05/31/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Early progression of chronic histologic lesions in kidney allografts represents the main finding in graft attrition. The objective of this retrospective cohort study was to elucidate whether HLA histocompatibility is associated with progression of chronic histologic lesions in the first year post-transplant. Established associations of de novo donor-specific antibody (dnDSA) formation with HLA mismatch and microvascular inflammation (MVI) were calculated to allow for comparability with other study cohorts. METHODS We included 117 adult kidney transplant recipients, transplanted between 2016 and 2020 from predominantly deceased donors, who had surveillance biopsies at 3 and 12 months. Histologic lesion scores were assessed according to the Banff classification. HLA mismatch scores [i.e. eplet, predicted indirectly recognizable HLA-epitopes algorithm (PIRCHE-II), HLA epitope mismatch algorithm (HLA-EMMA), HLA whole antigen A/B/DR] were calculated for all transplant pairs. Formation of dnDSAs was quantified by single antigen beads. RESULTS More than one-third of patients exhibited a progression of chronic lesion scores by at least one Banff grade in tubular atrophy (ct), interstitial fibrosis (ci), arteriolar hyalinosis (ah) and inflammation in the area of interstitial fibrosis and tubular atrophy (i-IFTA) from the 3- to the 12-month biopsy. Multivariable proportional odds logistic regression models revealed no association of HLA mismatch scores with progression of histologic lesions, except for ah and especially HLA-EMMA DRB1 [odds ratio (OR) = 1.10, 95% confidence interval (CI) 1.03-1.18]. Furthermore, the established associations of dnDSA formation with HLA mismatch and MVI (OR = 5.31, 95% CI 1.19-22.57) could be confirmed in our cohort. CONCLUSIONS These data support the association of HLA mismatch and alloimmune response, while suggesting that other factors contribute to early progression of chronic histologic lesions.
Collapse
Affiliation(s)
- Rhea Jabbour
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mariella G Gregorich
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS), Medical University of Vienna, Vienna, Austria
| | - Heinz Regele
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Johannes Kläger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Gottfried Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexander Kainz
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Jan U Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, Canada; Shared Health Services Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Tran J, Alrajhi I, Chang D, Sherwood KR, Keown P, Gill J, Kadatz M, Gill J, Lan JH. Clinical relevance of HLA-DQ eplet mismatch and maintenance immunosuppression with risk of allosensitization after kidney transplant failure. Front Genet 2024; 15:1383220. [PMID: 38638120 PMCID: PMC11024336 DOI: 10.3389/fgene.2024.1383220] [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: 02/07/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
The optimal immunosuppression management in patients with a failed kidney transplant remains uncertain. This study analyzed the association of class II HLA eplet mismatches and maintenance immunosuppression with allosensitization after graft failure in a well characterized cohort of 21 patients who failed a first kidney transplant. A clinically meaningful increase in cPRA in this study was defined as the cPRA that resulted in 50% reduction in the compatible donor pool measured from the time of transplant failure until the time of repeat transplantation, death, or end of study. The median cPRA at the time of failure was 12.13% (interquartile ranges = 0.00%, 83.72%) which increased to 62.76% (IQR = 4.34%, 99.18%) during the median follow-up of 27 (IQR = 18, 39) months. High HLA-DQ eplet mismatches were significantly associated with an increased risk of developing a clinically meaningful increase in cPRA (p = 0.02) and de novo DQ donor-specific antibody against the failed allograft (p = 0.02). We did not observe these associations in patients with high HLA-DR eplet mismatches. Most of the patients (88%) with a clinically meaningful increase in cPRA had both a high DQ eplet mismatch and a reduction in their immunosuppression, suggesting the association is modified by immunosuppression. The findings suggest HLA-DQ eplet mismatch analysis may serve as a useful tool to guide future clinical studies and trials which assess the management of immunosuppression in transplant failure patients who are repeat transplant candidates.
Collapse
Affiliation(s)
- Jenny Tran
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ibrahim Alrajhi
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Doris Chang
- Vancouver Coastal Health Research Institute, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Karen R. Sherwood
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul Keown
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health Research Institute, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jagbir Gill
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Providence Health Care Research Institute, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Kadatz
- Vancouver Coastal Health Research Institute, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John Gill
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Providence Health Care Research Institute, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - James H. Lan
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health Research Institute, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Rampersad C, Kong W, Wiebe C, Balshaw R, Bullard J, Villalobos APC, Trachtenberg A, Shaw J, Karpinski M, Nickerson PW, Ho J. Safety and efficacy of a reduced frequency viral monitoring strategy for Epstein-Barr virus, cytomegalovirus, and BK polyomavirus post-kidney transplant: A quality assurance initiative. Clin Transplant 2024; 38:e15292. [PMID: 38545888 DOI: 10.1111/ctr.15292] [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: 01/17/2024] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND There is variability in recommended viral monitoring protocols after kidney transplant. In response to increased demand for laboratory testing during the COVID-19 pandemic, the Transplant Manitoba Adult Kidney Program updated its monitoring protocols for cytomegalovirus (CMV), Epstein-Barr virus (EBV), and BK polyomavirus (BKV) to a reduced frequency. METHODS This single-center nested case-control study evaluated 252 adult kidney transplant recipients transplanted from 2015 to 2021, with the updated protocols effective on March 19th 2020. Cases included recipients transplanted after the protocol update who developed CMV, EBV, and BKV DNAemia and were matched to controls with DNAemia transplanted prior to the protocol update. The primary outcome was the difference in maximum DNA load titers between cases and matched controls. Secondary outcomes included time to initial DNAemia detection and DNAemia clearance. Safety outcomes of tissue-invasive viral disease were described. RESULTS There were 216 recipients transplanted preupdate and 36 recipients postupdate. There was no difference between cases and controls in maximum or first DNA load titers for EBV, CMV, or BKV. Cases experienced earlier EBV DNAemia detection (26 (IQR 8, 32) vs. 434 (IQR 96, 1184) days, p = .005). Median follow-up was significantly longer for recipients transplanted preupdate (4.3 vs. 1.3 years, p < .0001). After adjusting for follow-up time, there was no difference in DNAemia clearance or tissue-invasive viral disease. CONCLUSION Our findings suggest that reduced frequency viral monitoring protocols may be safe and cost-effective. This quality assurance initiative should be extended to detect longer-term and tissue-invasive disease outcomes.
Collapse
Affiliation(s)
- Christie Rampersad
- University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - William Kong
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chris Wiebe
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
| | - Robert Balshaw
- George and Fay Yee Centre for Healthcare Innovation, Winnipeg, Manitoba, Canada
| | - Jared Bullard
- Cadham Provincial Laboratory, Winnipeg, Manitoba, Canada
- Department of Pediatrics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Armelle Perez Cortes Villalobos
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
| | - Aaron Trachtenberg
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
| | - James Shaw
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
| | - Martin Karpinski
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
| | - Peter W Nickerson
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Julie Ho
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Transplant Manitoba Adult Kidney Program, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
17
|
Yanyiam P, Kantachuvesiri S, Thammanichanond D. Impact of HLA Eplet Mismatch on De Novo Donor Specific Antibody Formation After Kidney Transplantation. Transplant Proc 2024; 56:515-520. [PMID: 38368130 DOI: 10.1016/j.transproceed.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/16/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND HLA eplet mismatching is an alternative approach to assess the risk of developing de novo donor-specific antibodies (dnDSA) in kidney transplantation. This strategy may offer more precise risk stratification than conventional approaches. This study aimed to find the association between HLA eplet mismatches and dnDSA formation in Thai kidney transplant recipients. METHODS A retrospective cohort study of kidney transplant recipients transplanted between 2000 and 2021 at Ramathibodi Hospital was performed. Recipients with pretransplant panel reactive antibody >0% or without DSA testing post-transplant were excluded. One hundred fifty recipients were included in the final study. High-resolution HLA typing was imputed by the HaploStat application. HLA eplet mismatch analysis was conducted using HLAMatchmaker. The association between the number of eplet mismatches and the risk of dnDSA formation was assessed by Cox regression analysis. RESULTS Of 150 recipients, 43 were dnDSA-positive, and 107 were dnDSA-negative patients. Compared with the dnDSA-negative group, patients with class II dnDSA had significantly more HLA-DR/DQ antibody (Ab)-verified eplet mismatches (6 [IQR 4-8] vs 4 [IQR 1-7], P = .045). The receiver operating characteristics analysis showed that the HLA-DQ Ab-verified eplet mismatches ≥2 were the best predictive of HLA class II dnDSA development. The number of HLA-DQ Ab-verified eplet mismatches ≥2 had the highest hazard rate of HLA class II dnDSA occurrence (adjusted HR, 3.74; 95%CI, 1.24-11.24, P = .019). CONCLUSIONS HLA-DQ Ab-verified eplet mismatches are significantly associated with class II dnDSA development. Our data supports the utility of HLA eplet mismatching for donor-recipient risk assessment.
Collapse
Affiliation(s)
- Peerawit Yanyiam
- Histocompatibility and Immunogenetics Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Surasak Kantachuvesiri
- Division of Nephrology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Duangtawan Thammanichanond
- Histocompatibility and Immunogenetics Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
18
|
Heeger PS, Haro MC, Jordan S. Translating B cell immunology to the treatment of antibody-mediated allograft rejection. Nat Rev Nephrol 2024; 20:218-232. [PMID: 38168662 DOI: 10.1038/s41581-023-00791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 01/05/2024]
Abstract
Antibody-mediated rejection (AMR), including chronic AMR (cAMR), causes ~50% of kidney allograft losses each year. Despite attempts to develop well-tolerated and effective therapeutics for the management of AMR, to date, none has obtained FDA approval, thereby highlighting an urgent unmet medical need. Discoveries over the past decade from basic, translational and clinical studies of transplant recipients have provided a foundation for developing novel therapeutic approaches to preventing and treating AMR and cAMR. These interventions are aimed at reducing donor-specific antibody levels, decreasing graft injury and fibrosis, and preserving kidney function. Innovative approaches emerging from basic science findings include targeting interactions between alloreactive T cells and B cells, and depleting alloreactive memory B cells, as well as donor-specific antibody-producing plasmablasts and plasma cells. Therapies aimed at reducing the cytotoxic antibody effector functions mediated by natural killer cells and the complement system, and their associated pro-inflammatory cytokines, are also undergoing evaluation. The complexity of the pathogenesis of AMR and cAMR suggest that multiple approaches will probably be required to treat these disease processes effectively. Definitive answers await results from large, double-blind, multicentre, randomized controlled clinical trials.
Collapse
Affiliation(s)
- Peter S Heeger
- Comprehensive Transplant Center, Department of Medicine, Division of Nephrology Cedars-Sinai Medical Center Los Angeles, Los Angeles, CA, USA
| | - Maria Carrera Haro
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, Mount Sinai, NY, USA
| | - Stanley Jordan
- Comprehensive Transplant Center, Department of Medicine, Division of Nephrology Cedars-Sinai Medical Center Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Maguire C, Crivello P, Fleischhauer K, Isaacson D, Casillas A, Kramer CS, Copley HC, Heidt S, Kosmoliaptsis V, Meneghini M, Gmeiner M, Schold J, Louzoun Y, Tambur AR. Qualitative, rather than quantitative, differences between HLA-DQ alleles affect HLA-DQ immunogenicity in organ transplantation. HLA 2024; 103:e15455. [PMID: 38575370 PMCID: PMC11003724 DOI: 10.1111/tan.15455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
Prolonging the lifespan of transplanted organs is critical to combat the shortage of this life-saving resource. Chronic rejection, with irreversible demise of the allograft, is often caused by the development of donor-specific HLA antibodies. Currently, enumerating molecular (amino acid) mismatches between recipient and donor is promoted to identify patients at higher risk of developing HLA antibodies, for use in organ allocation, and immunosuppression-minimization strategies. We have counseled against the incorporation of such approaches into clinical use and hypothesized that not all molecular mismatches equally contribute to generation of donor-specific immune responses. Herein, we document statistical shortcomings in previous study design: for example, use of individuals who lack the ability to generate donor-specific-antibodies (HLA identical) as part of the negative cohort. We provide experimental evidence, using CRISPR-Cas9-edited cells, to rebut the claim that the HLAMatchmaker eplets represent "functional epitopes." We further used unique sub-cohorts of patients, those receiving an allograft with two HLA-DQ mismatches yet developing antibodies only to one mismatch (2MM1DSA), to interrogate differential immunogenicity. Our results demonstrate that mismatches of DQα05-heterodimers exhibit the highest immunogenicity. Additionally, we demonstrate that the DQα chain critically contributes to the overall qualities of DQ molecules. Lastly, our data proposes that an augmented risk to develop donor-specific HLA-DQ antibodies is dependent on qualitative (evolutionary and functional) divergence between recipient and donor, rather than the mere number of molecular mismatches. Overall, we propose an immunological mechanistic rationale to explain differential HLA-DQ immunogenicity, with potential ramifications for other pathological processes such as autoimmunity and infections.
Collapse
Affiliation(s)
- Chelsea Maguire
- Department of Surgery, Comprehensive Transplant Center; Northwestern University, Chicago, IL, USA. 60611
| | - Pietro Crivello
- Institute for Experimental Cellular Therapy, University Hospital Essen; Essen, Germany. 45127
| | - Katharina Fleischhauer
- Institute for Experimental Cellular Therapy, University Hospital Essen; Essen, Germany. 45127
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf; Essen, Germany. 45127
| | - Dylan Isaacson
- Department of Surgery, Comprehensive Transplant Center; Northwestern University, Chicago, IL, USA. 60611
| | - Aurora Casillas
- Department of Surgery, Comprehensive Transplant Center; Northwestern University, Chicago, IL, USA. 60611
| | - Cynthia S.M. Kramer
- Department of Immunology, Leiden University Medical Center; Netherlands. 2300-2334
| | - Hannah C. Copley
- Department of Surgery, University of Cambridge; Cambridge, United Kingdom. CB2 3AX
| | - Sebastian Heidt
- Department of Immunology, Leiden University Medical Center; Netherlands. 2300-2334
| | | | - Maria Meneghini
- Vall d’Hebron Institut de Recerca, Vall d’Hebron Hospital Universitari; Barcelona, Spain. 08035
| | - Michael Gmeiner
- Department of Economics, London School of Economics; London, United Kingdom. WC2A 2AE
| | - Jesse Schold
- Department of surgery, university of Colorado Anschutz Medical campus; Aurora, CO, USA. 80045
- Department of epidemiology, university of Colorado Anschutz Medical campus; Aurora, CO, USA. 80045
| | - Yoram Louzoun
- Department of mathematics Bar Ilan University; Ramat Gan, Israel. 5290002
| | - Anat R. Tambur
- Department of Surgery, Comprehensive Transplant Center; Northwestern University, Chicago, IL, USA. 60611
| |
Collapse
|
20
|
Senev A, Tambur AR, Kosmoliaptsis V, Copley HC, García-Sánchez C, Usenko C, Ildstad ST, Leventhal JR. HLA molecular mismatches and induced donor-specific tolerance in combined living donor kidney and hematopoietic stem cell transplantation. Front Immunol 2024; 15:1377535. [PMID: 38601147 PMCID: PMC11004438 DOI: 10.3389/fimmu.2024.1377535] [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: 01/27/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction We investigated the potential role of HLA molecular mismatches (MM) in achieving stable chimerism, allowing for donor-specific tolerance in patients undergoing combined living donor kidney and hematopoietic stem cell transplantation (HSCT). Methods All patients with available DNA samples (N=32) who participated in a phase 2 clinical trial (NCT00498160) where they received an HLA mismatched co-transplantation of living donor kidney and facilitating cell-enriched HSCT were included in this study. High-resolution HLA genotyping data were used to calculate HLA amino acid mismatches (AAMM), Eplet MM, three-dimensional electrostatic mismatch scores (EMS-3D), PIRCHE scores, HLA-DPB1 T-cell epitope group MM, HLA-B leader sequence MM, and KIR ligands MM between the donor and recipient in both directions. HLA MM were analyzed to test for correlation with the development of chimerism, graft vs. host disease (GvHD), de novo DSA, and graft rejection. Results Follow-up time of this cohort was 6-13.5 years. Of the 32 patients, 26 developed high-level donor or mixed stable chimerism, followed by complete withdrawal of immunosuppression (IS) in 25 patients. The remaining six of the 32 patients had transient chimerism or no engraftment and were maintained on IS (On-IS). In host versus graft direction, a trend toward higher median number of HLA-DRB1 MM scores was seen in patients On-IS compared to patients with high-level donor/mixed chimerism, using any of the HLA MM modalities; however, initial statistical significance was observed only for the EMS-3D score (0.45 [IQR, 0.30-0.61] vs. 0.24 [IQR, 0.18-0.36], respectively; p=0.036), which was lost when applying the Bonferroni correction. No statistically significant differences between the two groups were observed for AAMM, EMS-3D, Eplet MM, and PIRCHE-II scores calculated in graft versus host direction. No associations were found between development of chimerism and GvHD and non-permissive HLA-DPB1 T-cell epitope group MM, HLA-B leader sequence, and KIR ligands MM. Conclusion Our results suggest an association between HLA-DRB1 molecular mismatches and achieving stable chimerism, particularly when electrostatic quality of the mismatch is considered. The non-permissive HLA-DPB1 T-cell epitope group, HLA-B leader sequence, and KIR ligands MM do not predict chimerism and GvHD in this combined kidney/HSCT transplant patient cohort. Further work is needed to validate our findings. Clinical trial registration https://clinicaltrials.gov/study/NCT00498160, identifier NCT00498160.
Collapse
Affiliation(s)
- Aleksandar Senev
- The Comprehensive Transplant Center (CTC) at Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Anat R. Tambur
- The Comprehensive Transplant Center (CTC) at Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Vasilis Kosmoliaptsis
- Department of Surgery, Addenbrooke’s Hospital, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Hannah Charlotte Copley
- Department of Surgery, Addenbrooke’s Hospital, University of Cambridge and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Cynthia García-Sánchez
- The Comprehensive Transplant Center (CTC) at Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Crystal Usenko
- The Comprehensive Transplant Center (CTC) at Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Suzanne T. Ildstad
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY, United States
| | - Joseph R. Leventhal
- The Comprehensive Transplant Center (CTC) at Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
21
|
Meneghini M, Tambur AR. HLA (emphasis on DQ) compatibility for longer allograft survival in pediatric transplantation: Modern evidence and challenges. Pediatr Transplant 2024; 28:e14606. [PMID: 37716000 DOI: 10.1111/petr.14606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/08/2023] [Accepted: 08/28/2023] [Indexed: 09/18/2023]
Abstract
Kidney transplantation is the treatment of choice for children with end-stage kidney failure, yet suboptimal outcomes, the need for long-term immunosuppression, and the dependency on consecutive transplants pose significant barriers to success. Providing better HLA-matched organs to pediatric patients seems to be the most logical approach to improve graft and patient outcomes and to reduce risk of anti-HLA sensitization after graft failure. We here review recent literature on HLA matching in pediatric kidney transplantation. We further review newer approaches attempting to improve matching by using molecular mismatch load analysis. Our main focus is on the role of HLA-DQ compatibility between recipient and donor. We further emphasize the need to develop creative approaches that will support HLA (and DQ) matching utilization in organ allocation schemes, at least in those geared specifically for pediatric patients.
Collapse
Affiliation(s)
- Maria Meneghini
- Transplant Immunology Laboratory, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
- Nephrology and Kidney Transplantation Department, Vall d'Hebron University Hospital, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Anat Roitberg Tambur
- Transplant Immunology Laboratory, Feinberg School of Medicine, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
22
|
Zhang Q, Locke AF, Alvarez AC, Cabarong ML, Liv LC, Alfaro BGP, Gjertson DW, Reed EF. Advancing precision in histocompatibility and immunogenetics: a comprehensive review of the UCLA exchange program. Front Genet 2024; 15:1352764. [PMID: 38362203 PMCID: PMC10867271 DOI: 10.3389/fgene.2024.1352764] [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: 12/08/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024] Open
Abstract
Precise typing of human leukocyte antigens (HLA) is crucial for clinical hematopoietic stem cell and solid organ transplantations, transfusion medicine, HLA-related disease association, and drug hypersensitivity analysis. The UCLA Cell Exchange program has played a vital role in providing educational and proficiency testing surveys to HLA laboratories worldwide for the past 5 decades. This article highlights the significant contribution of the UCLA Cell and DNA Exchange Programs in advancing HLA antibody testing, genotyping, crossmatches, and, more recently, virtual crossmatches. Additionally, we discuss future directions of the UCLA Cell Exchange program to support histocompatibility testing to adapt to the fast-evolving field of immunotherapy, tolerance and xenotransplantation.
Collapse
Affiliation(s)
- Qiuheng Zhang
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Johnson AC, Zhang J, Karadkhele G, Gragert L, Hertzberg V, Larsen CP. Belatacept with time-limited tacrolimus coimmunosuppression modifies the 3-year risk of eplet mismatch in kidney transplantation. Am J Transplant 2024; 24:260-270. [PMID: 37778459 PMCID: PMC10842047 DOI: 10.1016/j.ajt.2023.09.011] [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: 01/04/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Solid organ transplant donor-recipient eplet mismatch has been correlated with donor-specific antibody (DSA) formation, antibody-mediated rejection, and overall rejection rates. However, studies have been predominantly in patients on tacrolimus-based immunosuppression regimens and have not fully explored differences in ethnically and racially diverse populations. Evidence indicates that patients on belatacept have lower rates of DSA formation, suggesting mediation of the immunogenicity of mismatched human leukocyte antigen polymorphisms. We performed a retrospective, single-center analysis of class II eplet disparity in a cohort of kidney transplant recipients treated using belatacept with tacrolimus induction (Bela/TacTL) or tacrolimus regimens between 2016 and 2019. Bela/TacTL (n = 294) and tacrolimus (n = 294) cohorts were propensity score-matched with standardized difference <0.15. Single-molecule eplet risk level was associated with immune event rates for both groups. In Cox regression analysis stratified by eplet risk level, Bela/TacTL immunosuppression was associated with a decreased rate of DSA (hazard ratio [HR] = 0.4), antibody-mediated rejection (HR = 0.2), and rejection (HR = 0.45). In the low-risk group, cumulative graft failure was lower for patients on Bela/TacTL (P < .02). Analysis of eplet mismatch burden may be a useful adjunct in identifying high-risk populations with increased immunosuppression requirements and should encourage the design of allocation rules to incentivize lower-risk pairings without negatively impacting equity in access.
Collapse
Affiliation(s)
- Aileen C Johnson
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Joan Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Geeta Karadkhele
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Loren Gragert
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Vicki Hertzberg
- Woodruff School of Nursing, Emory University, Atlanta, Georgia, USA
| | - Christian P Larsen
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
24
|
Yang BQ, Bai YJ, Wang LL, Dai B, Li YM, Tao Y, Shi YY. The impact of pretransplant suspected HLA antibody on the long-term outcome of the graft kidney: A retrospective cohort study. Transpl Immunol 2024; 82:101922. [PMID: 37657691 DOI: 10.1016/j.trim.2023.101922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/16/2023] [Accepted: 08/27/2023] [Indexed: 09/03/2023]
Abstract
INTRODUCTION The preoperative examination of kidney transplantation includes HLA antibody screening to initially determine the presence of preexisting donor-specific antibody (DSA) that mediates hyperacute rejection. Recipients with positive HLA antibodies require further HLA specificity analysis to type the antigen and determine the antigen mismatches between the donor and recipient. However, recipients with suspected antibodies would have no further HLA specificity analysis. It is unclear whether suspected HLA antibodies would affect renal graft function. This study aimed to explore the impact of pretransplant suspected HLA antibody on the long-term outcome of the graft kidney and thus determine the necessity of routinely performing the HLA specificity analysis in recipients with suspected HLA antibodies preoperatively. METHODS This is a single-center retrospective cohort study. 179 kidney transplant recipients (KTRs) were included and further divided into HLA antibody-negative group (Group 1) and HLA antibody-suspected groups (Group 2) based on the result of the pretransplant HLA antibody screen test. And the antibody-suspected group was further divided into a low-mismatched group (Group A) and a high-mismatched group (Group B) according to the HLA specificity analysis. We tracked the renal function indexes, biochemical indexes, and posttransplant adverse events within 5 years after transplantation and explored the necessity of further HLA specificity analysis in recipients with pretransplant suspected HLA antibodies. RESULTS There was no statistically significant difference in demographics between HLA antibody-negative group and HLA antibody-suspected groups. At 5 years of follow-up, the KTRs in HLA antibody-negative group had significantly higher eGFR levels, lower serum creatinine levels, and less urinary protein compared to those in antibody-suspected group. Meanwhile, the KTRs in low-mismatched group also had significantly higher eGFR levels, lower serum creatinine levels, and less proteinuria compared to those in high-mismatched group. Correlation analysis showed that the age of KTRs, urinary protein levels and the load capacity of HLA mismatches were associated with eGFR levels of KTRs at 5 year posttransplant. CONCLUSION KTRs with suspected HLA antibodies before kidney transplantation have worse graft function than the preoperative HLA antibody-negative recipients in the long-term posttransplant follow-up. The specific load capacity of HLA mismatches, the age of the recipient and the urinary protein was found to be negatively correlated with long-term posttransplant renal outcomes. It is necessary to undergo further HLA specificity analysis for recipients with suspected HLA antibodies in HLA antibody screen test to explicit HLA mismatches and improve long-term posttransplant outcomes.
Collapse
Affiliation(s)
- Bin-Qi Yang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yang-Juan Bai
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu 610041,China
| | - Lan-Lan Wang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu 610041,China
| | - Bo Dai
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu 610041,China
| | - Ya-Mei Li
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu 610041,China
| | - Ye Tao
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yun-Ying Shi
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
25
|
Kramer CSM, Bezstarosti S, Franke-van Dijk MEI, Vergunst M, Roelen DL, Uyar-Mercankaya M, Voogt-Bakker KH, Heidt S. Antibody verification of HLA class I and class II eplets by human monoclonal HLA antibodies. HLA 2024; 103:e15345. [PMID: 38239050 DOI: 10.1111/tan.15345] [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: 09/08/2023] [Revised: 12/11/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024]
Abstract
In solid organ transplantation, formation of de novo donor-specific HLA antibodies is induced by mismatched eplets on donor HLA molecules. While several studies have shown a strong correlation between the number of eplet mismatches and inferior outcomes, not every eplet mismatch is immunogenic. Eplets are theoretically defined entities, necessitating formal proof that they can be recognised and bound by antibodies. This antibody verification is pivotal to ensure that clinically relevant eplets are considered in studies on molecular matching. Recombinant human HLA-specific monoclonal antibodies (mAbs) were generated from HLA-reactive B cell clones isolated from HLA immunised individuals using recombinant HLA molecules. Subsequently, the reactivity patterns of the mAbs obtained from single antigen bead assay were analysed using HLA-EMMA software to identify single or configurations of solvent accessible amino acids uniquely present on the reactive HLA alleles and were mapped to eplets. Two HLA class I and seven HLA class II-specific human mAbs were generated from four individuals. Extensive mAb reactivity analysis, led to antibody verification of three HLA-DR-specific eplets, and conversion of five eplets (one HLA-A, one HLA-B, two HLA-DR, and one HLA-DP), from provisionally verified to truly antibody-verified. Finally, one HLA-DQ-specific eplet was upgraded from level A2 to level A1 verification evidence. The generation of recombinant human HLA-specific mAbs with different specificities contributes significantly to the antibody verification of eplets and therefore is instrumental for implementation of eplet matching in the clinical setting.
Collapse
Affiliation(s)
- Cynthia S M Kramer
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne Bezstarosti
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Manon Vergunst
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Kim H Voogt-Bakker
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Schapranow MP, Bayat M, Rasheed A, Naik M, Graf V, Schmidt D, Budde K, Cardinal H, Sapir-Pichhadze R, Fenninger F, Sherwood K, Keown P, Günther OP, Pandl KD, Leiser F, Thiebes S, Sunyaev A, Niemann M, Schimanski A, Klein T. NephroCAGE-German-Canadian Consortium on AI for Improved Kidney Transplantation Outcome: Protocol for an Algorithm Development and Validation Study. JMIR Res Protoc 2023; 12:e48892. [PMID: 38133915 PMCID: PMC10770792 DOI: 10.2196/48892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Recent advances in hardware and software enabled the use of artificial intelligence (AI) algorithms for analysis of complex data in a wide range of daily-life use cases. We aim to explore the benefits of applying AI to a specific use case in transplant nephrology: risk prediction for severe posttransplant events. For the first time, we combine multinational real-world transplant data, which require specific legal and technical protection measures. OBJECTIVE The German-Canadian NephroCAGE consortium aims to develop and evaluate specific processes, software tools, and methods to (1) combine transplant data of more than 8000 cases over the past decades from leading transplant centers in Germany and Canada, (2) implement specific measures to protect sensitive transplant data, and (3) use multinational data as a foundation for developing high-quality prognostic AI models. METHODS To protect sensitive transplant data addressing the first and second objectives, we aim to implement a decentralized NephroCAGE federated learning infrastructure upon a private blockchain. Our NephroCAGE federated learning infrastructure enables a switch of paradigms: instead of pooling sensitive data into a central database for analysis, it enables the transfer of clinical prediction models (CPMs) to clinical sites for local data analyses. Thus, sensitive transplant data reside protected in their original sites while the comparable small algorithms are exchanged instead. For our third objective, we will compare the performance of selected AI algorithms, for example, random forest and extreme gradient boosting, as foundation for CPMs to predict severe short- and long-term posttransplant risks, for example, graft failure or mortality. The CPMs will be trained on donor and recipient data from retrospective cohorts of kidney transplant patients. RESULTS We have received initial funding for NephroCAGE in February 2021. All clinical partners have applied for and received ethics approval as of 2022. The process of exploration of clinical transplant database for variable extraction has started at all the centers in 2022. In total, 8120 patient records have been retrieved as of August 2023. The development and validation of CPMs is ongoing as of 2023. CONCLUSIONS For the first time, we will (1) combine kidney transplant data from nephrology centers in Germany and Canada, (2) implement federated learning as a foundation to use such real-world transplant data as a basis for the training of CPMs in a privacy-preserving way, and (3) develop a learning software system to investigate population specifics, for example, to understand population heterogeneity, treatment specificities, and individual impact on selected posttransplant outcomes. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/48892.
Collapse
Affiliation(s)
- Matthieu-P Schapranow
- Hasso Plattner Institute for Digital Engineering, University of Potsdam, Potsdam, Germany
| | - Mozhgan Bayat
- Hasso Plattner Institute for Digital Engineering, University of Potsdam, Potsdam, Germany
| | - Aadil Rasheed
- Hasso Plattner Institute for Digital Engineering, University of Potsdam, Potsdam, Germany
| | - Marcel Naik
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Verena Graf
- Geschäftsbereich IT, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Danilo Schmidt
- Geschäftsbereich IT, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Héloïse Cardinal
- Research Centre, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Ruth Sapir-Pichhadze
- Division of Nephrology and Multi-Organ Transplant Program, Department of Medicine and Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Franz Fenninger
- Division of Nephrology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Karen Sherwood
- Division of Nephrology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul Keown
- Division of Nephrology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Konstantin D Pandl
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Florian Leiser
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Scott Thiebes
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ali Sunyaev
- Department of Economics and Management, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | | | | | | |
Collapse
|
27
|
Betjes MGH, Kal-van Gestel J, Roodnat JI, de Weerd AE. The Incidence of Antibody-Mediated Rejection Is Age-Related, Plateaus Late After Kidney Transplantation, and Contributes Little to Graft Loss in the Older Recipients. Transpl Int 2023; 36:11751. [PMID: 38188697 PMCID: PMC10768842 DOI: 10.3389/ti.2023.11751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
It is not known whether antibody-mediated rejection (ABMR) is age-related, whether it plateaus late after transplantation, and to what extent it contributes to graft loss in older recipients. Patients transplanted between 2010 and 2015 (n = 1,054) in a single center had regular follow-up until January 2023. Recipients were divided into age groups at transplantation: 18-39 years ("young"), 40-55 years ("middle age"), and >55 years ("elderly"). Ten years after transplantation the cumulative % of recipients with ABMR was 17% in young, 15% in middle age, and 12% in elderly recipients (p < 0.001). The cumulative incidence of ABMR increased over time and plateaued 8-10 years after transplantation. In the elderly, with a median follow-up of 7.5 years, on average 30% of the recipients with ABMR died with a functional graft and ABMR contributed only 4% to overall graft loss in this group. These results were cross-validated in a cohort of recipients with >15 years follow-up. Multivariate cox-regression analysis showed that increasing recipient age was independently associated with decreasing risk for ABMR. In conclusion, the cumulative risk for ABMR is age-dependent, plateaus late after transplantation, and contributes little to overall graft loss in older recipients.
Collapse
Affiliation(s)
- Michiel G. H. Betjes
- Rotterdam Transplantation Institute, Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
28
|
Wiebe C, Balshaw R, Gibson IW, Ho J, Shaw J, Karpinski M, Trachtenberg A, Pochinco D, Goldberg A, Birk P, Pinsk M, Rush DN, Nickerson PW. A rational approach to guide cost-effective de novo donor-specific antibody surveillance with tacrolimus immunosuppression. Am J Transplant 2023; 23:1882-1892. [PMID: 37543094 DOI: 10.1016/j.ajt.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
De novo donor-specific antibody (dnDSA) after renal transplantation has been shown to correlate with antibody-mediated rejection and allograft loss. However, the lack of proven interventions and the time and cost associated with annual screening for dnDSA are difficult to justify for all recipients. We studied a well-characterized consecutive cohort (n = 949) with over 15 years of prospective dnDSA surveillance to identify risk factors that would help institute a resource-responsible surveillance strategy. Younger recipient age and HLA-DR/DQ molecular mismatch were independent predictors of dnDSA development. Combining both risk factors into recipient age molecular mismatch categories, we found that 52% of recipients could be categorized as low-risk for dnDSA development (median subclinical dnDSA-free survival at 5 and 10 years, 98% and 97%, respectively). After adjustment, multivariate correlates of dnDSA development included tacrolimus versus cyclosporin maintenance immunosuppression (hazard ratio [HR], 0.37; 95% CI, 0.2-0.6; P < .0001) and recipient age molecular mismatch category: intermediate versus low (HR, 2.48; 95% CI, 1.5-4.2; P = .0007), high versus intermediate (HR, 2.56; 95% CI, 1.6-4.2; P = .0002), and high versus low (HR, 6.36; 95% CI, 3.7-10.8; P < .00001). When combined, recipient age and HLA-DR/DQ molecular mismatch provide a novel data-driven approach to reduce testing by >50% while selecting those most likely to benefit from dnDSA surveillance.
Collapse
Affiliation(s)
- Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Shared Health Services Manitoba, Winnipeg, Manitoba, Canada.
| | - Rob Balshaw
- George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ian W Gibson
- Shared Health Services Manitoba, Winnipeg, Manitoba, Canada; Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Julie Ho
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Shared Health Services Manitoba, Winnipeg, Manitoba, Canada
| | - Jamie Shaw
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Martin Karpinski
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Aaron Trachtenberg
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Aviva Goldberg
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Patricia Birk
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maury Pinsk
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David N Rush
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Shared Health Services Manitoba, Winnipeg, Manitoba, Canada
| | - Peter W Nickerson
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Shared Health Services Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
29
|
Gao S, Gong H, Li M, Lan P, Zhang M, Kuang P, Zhang Y, Hu X, Ding C, Li Y, Ding X, Xue W, Zheng J. HLA B eplet mismatches in the context of delayed graft function and low tacrolimus trough levels are risk factors influencing the generation of de novo donor-specific antibodies and acute rejection in the early stage after kidney transplantation. Transpl Immunol 2023; 81:101955. [PMID: 37931666 DOI: 10.1016/j.trim.2023.101955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND De novo donor-specific antibody (dnDSA) generation and acute rejection (AR) are the main factors affecting long-term graft survival. This study aims to investigate human leukocyte antigen (HLA) eplet mismatching (MM), delayed graft function (DGF), and tacrolimus (TAC) trough levels on the occurrence of dnDSA and AR in the early stages after kidney transplantation (KT). METHODS This retrospective study included 526 cases of deceased donation KT. The effects of DGF, HLA eplet MM, and TAC trough levels on dnDSA and AR occurrence were analyzed with logistic regression analysis. RESULTS Multivariate logistic regression analysis showed the independent risk factor of dnDSA generation was HLA B eplet MM (OR: 1.201, 95% CI: 1.007-1.431, P = 0.041). The independent risk factors of AR occurrence include DGF (OR: 4.045, 95% CI: 1.047-15.626, P = 0.043), HLA B eplet MM (OR: 1.090, 95% CI: 1.000-1.187, P = 0.050), and TAC trough levels at 12 months (OR: 0.750, 95% CI: 565-0.997, P = 0.048). HLA B eplet MM combined with DGF and TAC trough levels at 12 months increased the predictive value of dnDSA (AUC 0.735) and AR (AUC 0.730) occurrence. HLA B eplet MM > 9 and TAC trough levels below 5.95 ng/mL at 12 months could increase the risk of early AR occurrence. CONCLUSIONS HLA B eplet MM, DGF, and TAC trough levels at 12 months after KT could affect the occurrence of dnDSA and AR in the early stage of KT.
Collapse
Affiliation(s)
- Shan Gao
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Huilin Gong
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Meihe Li
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ping Lan
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Minyue Zhang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Peidan Kuang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ying Zhang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaojun Hu
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chenguang Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yang Li
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wujun Xue
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
30
|
Alves TA, Nascimento E, de Castro LB, Fabreti-Oliveira RA. Impact of HLA eplet mismatch load in immunological outcomes after living donor kidney transplantation. Transpl Immunol 2023; 80:101908. [PMID: 37536379 DOI: 10.1016/j.trim.2023.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/05/2023]
Abstract
INTRODUCTION HLA eplets mismatches (eMM) have been associated with negative kidney outcomes after transplantation, such as the development of de novo donor-specific antibody (dnDSA), antibody-mediated rejection (ABMR), and early graft loss. This study aimed to evaluate the clinical effects of the HLA eMM load on dnDSA development, ABMR, renal function, allograft survival and graft loss. MATERIAL AND METHODS This retrospective study involved 159 living donor kidney transplant patients categorized into groups based on antigen HLA mismatches assessed traditionally and HLA eMM load. Patients had followed for at least one year. The EpViX online program was used to evaluate the HLA eMM load. Cox models were constructed to assess the risk of graft loss. Kaplan-Meier survival curves were carried out. The analyses had performed using the R program and p < 0.05 was considered significant. RESULTS From all 159 patients, 28 (17.6%) lost their allografts. Rejection episodes occurred in 37.1% of patients, 13.6% of whom were ABMR. Patients with rejection episodes had higher HLA-AB (p = 0.032) and HLA-DR (p = 0.008) HLA eMM load, HLA-AB (p = 0.006) and HLA-DR (p = 0.009) antigens mismatches, and higher proportions of the following eMM in the HLA-DR locus: 70R eMM (p = 0.015), 70RE (p = 0.015), 74E (p = 0.015) and 48Q (p = 0.047). In multiple models, the presence of HLA-DR 70qq eMM (HR 3.75, 95% CI 1.47; 9.55) add an increase in creatinine levels at 1-year (HR 3.87, 95% CI 2.30, 6.53) were associated with the risk of graft loss. CONCLUSION The HLA eMM load was related to episodes of rejection and allograft loss. The HLA-DR eMM was most strongly associated with a worse immunologic outcome than eMM mismatches for HLA-AB.
Collapse
Affiliation(s)
- Thiago Abramo Alves
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; University Hospital of the Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil.
| | - Evaldo Nascimento
- IMUNOLAB - Laboratory of Histocompatibility, Belo Horizonte, Minas Gerais, Brazil; Institute of Research and Education of the Hospital Santa Casa, Belo Horizonte, Minas Gerais, Brazil.
| | | | - Raquel Aparecida Fabreti-Oliveira
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; IMUNOLAB - Laboratory of Histocompatibility, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
31
|
Hu Q, Christian D, Yang G, Burns C, Berka N. Identification of four novel HLA-DQ alleles, HLA-DQA1*01:106, -DQA1*01:107, -DQA1*05:74 and -DQB1*05:01:48. HLA 2023; 102:254-256. [PMID: 37191269 DOI: 10.1111/tan.15076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023]
Abstract
Characterization of new alleles, three HLA-DQA1 and one HLA-DQB1, bearing non-synonymous and synonymous mutations, respectively.
Collapse
Affiliation(s)
- Qian Hu
- Histocompatibility and Immunogenetics Laboratory, Alberta Precision Laboratories, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Danielle Christian
- Histocompatibility and Immunogenetics Laboratory, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Guang Yang
- Histocompatibility and Immunogenetics Laboratory, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Caitlyn Burns
- Histocompatibility and Immunogenetics Laboratory, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Noureddine Berka
- Histocompatibility and Immunogenetics Laboratory, Alberta Precision Laboratories, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Devriese M, Usureau C, Lion J, Sayegh C, Carmagnat M, Caillat-Zucman S, Mooney N, Lemonnier F, Taupin JL. Assessing the allogenic realness of the Cw1/12/15 pattern occurring in the LABScreen single antigen assay. HLA 2023; 102:157-167. [PMID: 37067656 DOI: 10.1111/tan.15025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 04/18/2023]
Abstract
Several technical limitations of Luminex single antigen (LSA) assays have been described so far. This study focused on a reactivity pattern observed in many sera that cannot be explained by eplets described in the Epitope Registry database and sometimes appearing against a self-HLA allele or antigen. In most cases, this pattern is revealed by a discrepant result when compared with other assays (Luminex PRA, cell-binding assays such as flow cytometry cross match, LSA from another manufacturer…). We focus here on the Cw1/12/15 pattern appearing on the LABScreen class I LSA provided by One Lambda. We documented its behavior using this LSA after acid denaturation of the beads, using Lifecodes LSA from Immucor, and adsorption of sera either on spleen mononuclear cells from deceased donors or on single HLA transfected cell clones. We studied 33 sera from different patients positive for the three Cw beads, selected from our routine patients' LSA database. Nine patients had transplants from a Cw12 or Cw15 donor without any pejorative evolution of the graft, nor post-transplant MFI (mean fluorescence intensity) increase of the Cw1/12/15 beads. A significant increase of MFI was observed after acid denaturation of the LABScreen beads. All sera tested by Lifecodes LSA were negative for these Cw beads. Finally, we found no significant difference of MFI after adsorption on cells from either origin. Therefore, the Cw1/12/15 pattern appears to be a false positive reactivity of the LABScreen single antigen assay.
Collapse
Affiliation(s)
- Magali Devriese
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Cédric Usureau
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Julien Lion
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Caroline Sayegh
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Maryvonnick Carmagnat
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Sophie Caillat-Zucman
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Nuala Mooney
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | | | - Jean Luc Taupin
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint Louis, Paris, France
- INSERM UMR 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| |
Collapse
|
33
|
Edwards L, Bentley C, Burgess M, Sapir-Pichhadze R, Hartell D, Keown P, Bryan S. Adding epitope compatibility to deceased donor kidney allocation criteria: recommendations from a pan-Canadian online public deliberation. BMC Nephrol 2023; 24:165. [PMID: 37296384 PMCID: PMC10255937 DOI: 10.1186/s12882-023-03224-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND The widening supply-demand imbalance for kidneys necessitates finding ways to reduce rejection and improve transplant outcomes. Human leukocyte antigen (HLA) epitope compatibility between donor and recipient may minimize premature graft loss and prolong survival, but incorporating this strategy to deceased donor allocation criteria prioritizes transplant outcomes over wait times. An online public deliberation was held to identify acceptable trade-offs when implementing epitope compatibility to guide Canadian policymakers and health professionals in deciding how best to allocate kidneys fairly. METHODS Invitations were mailed to 35,000 randomly-selected Canadian households, with over-sampling of rural/remote locations. Participants were selected for socio-demographic diversity and geographic representation. Five two-hour online sessions were held from November-December 2021. Participants received an information booklet and heard from expert speakers prior to deliberating on how to fairly implement epitope compatibility for transplant candidates and governance issues. Participants collectively generated and voted on recommendations. In the final session, kidney donation and allocation policymakers engaged with participants. Sessions were recorded and transcribed. RESULTS Thirty-two individuals participated and generated nine recommendations. There was consensus on adding epitope compatibility to the existing deceased donor kidney allocation criteria. However, participants recommended including safeguards/flexibility around this (e.g., mitigating declining health). They called for a transition period to epitope compatibility, including an ongoing comprehensive public education program. Participants unanimously recommended regular monitoring and public sharing of epitope-based transplant outcomes. CONCLUSIONS Participants supported adding epitope compatibility to kidney allocation criteria, but advised safeguards and flexibility around implementation. These recommendations provide guidance to policymakers about incorporating epitope-based deceased donor allocation criteria.
Collapse
Affiliation(s)
- Louisa Edwards
- School of Population & Public Health, University of British Columbia (UBC), 717 - 828 West 10th Avenue, Research Pavilion, Vancouver, BC, V5Z 1M9, Canada.
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, Canada.
| | | | - Michael Burgess
- School of Population & Public Health, University of British Columbia (UBC), 717 - 828 West 10th Avenue, Research Pavilion, Vancouver, BC, V5Z 1M9, Canada
- W. Maurice Young Centre for Applied Ethics, UBC, Vancouver, Canada
| | - Ruth Sapir-Pichhadze
- Division of Nephrology, Department of Medicine, McGill University, Montreal, Canada
- Centre for Outcomes Research & Evaluation, Research Institute of the McGill University Health Centre, Vancouver, Canada
| | | | - Paul Keown
- Department of Medicine, UBC, Vancouver, Canada
- Immune Centre of BC, Vancouver Coastal Health, Vancouver, Canada
| | - Stirling Bryan
- School of Population & Public Health, University of British Columbia (UBC), 717 - 828 West 10th Avenue, Research Pavilion, Vancouver, BC, V5Z 1M9, Canada
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, Canada
| |
Collapse
|
34
|
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
|
35
|
Franco-Acevedo A, Comes J, Mack JJ, Valenzuela NM. New insights into maladaptive vascular responses to donor specific HLA antibodies in organ transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1146040. [PMID: 38993843 PMCID: PMC11235244 DOI: 10.3389/frtra.2023.1146040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 07/13/2024]
Abstract
Transplant vasculopathy (TV) causes thickening of donor blood vessels in transplanted organs, and is a significant cause of graft loss and mortality in allograft recipients. It is known that patients with repeated acute rejection and/or donor specific antibodies are predisposed to TV. Nevertheless, the exact molecular mechanisms by which alloimmune injury culminates in this disease have not been fully delineated. As a result of this incomplete knowledge, there is currently a lack of effective therapies for this disease. The immediate intracellular signaling and the acute effects elicited by anti-donor HLA antibodies are well-described and continuing to be revealed in deeper detail. Further, advances in rejection diagnostics, including intragraft gene expression, provide clues to the inflammatory changes within allografts. However, mechanisms linking these events with long-term outcomes, particularly the maladaptive vascular remodeling seen in transplant vasculopathy, are still being delineated. New evidence demonstrates alterations in non-coding RNA profiles and the occurrence of endothelial to mesenchymal transition (EndMT) during acute antibody-mediated graft injury. EndMT is also readily apparent in numerous settings of non-transplant intimal hyperplasia, and lessons can be learned from advances in those fields. This review will provide an update on these recent developments and remaining questions in our understanding of HLA antibody-induced vascular damage, framed within a broader consideration of manifestations and implications across transplanted organ types.
Collapse
Affiliation(s)
- Adriana Franco-Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Johanna Comes
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA, United States
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
36
|
Kim JJ, Fichtner A, Copley HC, Gragert L, Süsal C, Dello Strologo L, Oh J, Pape L, Weber LT, Weitz M, König J, Krupka K, Tönshoff B, Kosmoliaptsis V. Molecular HLA mismatching for prediction of primary humoral alloimmunity and graft function deterioration in paediatric kidney transplantation. Front Immunol 2023; 14:1092335. [PMID: 37033962 PMCID: PMC10080391 DOI: 10.3389/fimmu.2023.1092335] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
Introduction Rejection remains the main cause of allograft failure in paediatric kidney transplantation and is driven by donor-recipient HLA mismatching. Modern computational algorithms enable assessment of HLA mismatch immunogenicity at the molecular level (molecular-mismatch, molMM). Whilst molMM has been shown to correlate with alloimmune outcomes, evidence demonstrating improved prediction performance against traditional antigen mismatching (antMM) is lacking. Methods We analysed 177 patients from the CERTAIN registry (median follow-up 4.5 years). molMM scores included Amino-Acid-Mismatch-Score (AAMS), Electrostatic-Mismatch-Score (EMS3D) and netMHCIIpan (netMHC1k: peptide binding affinity ≤1000 nM; netMHC: binding affinity ≤500 nM plus rank <2%). We stratified patients into high/low-risk groups based on risk models of DSA development. Results Donor-specific HLA antibodies (DSA) predominantly targeted the highest scoring molMM donor antigen within each HLA locus. MolMM scores offered superior discrimination versus antMM in predicting de novo DSA for all HLA loci; the EMS3D algorithm had particularly consistent performance (area under the receiver operating characteristic curve (AUC) >0.7 for all HLA loci vs. 0.52-0.70 for antMM). ABMR (but not TCMR) was associated with HLA-DQ molMM scores (AAMS, EMS3D and netMHC). Patients with high-risk HLA-DQ molMM had increased risk of graft function deterioration (50% reduction in baseline eGFR (eGFR50), adjusted HR: 3.5, 95% CI 1.6-8.2 high vs. low EMS3D). Multivariable modelling of the eGFR50 outcome using EMS3D HLA-DQ stratification showed better discrimination (AUC EMS3D vs. antMM at 2 years: 0.81 vs. 0.77, at 4.5 years: 0.72 vs. 0.64) and stratified more patients into the low-risk group, compared to traditional antMM. Conclusion Molecular mismatching was superior to antigen mismatching in predicting humoral alloimmunity. Molecular HLA-DQ mismatching appears to be a significant prognostic factor for graft function deterioration in paediatric kidney transplantation.
Collapse
Affiliation(s)
- Jon Jin Kim
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
- Department of Paediatric Nephrology, Nottingham University Hospital, Nottingham, United Kingdom
| | - Alexander Fichtner
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Hannah C. Copley
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Loren Gragert
- School of Medicine, Tulane University, New Orleans, LA, United States
| | - Caner Süsal
- Transplantation Immunology, Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Jun Oh
- University Hospital Hamburg, Pediatric Nephrology, Hamburg, Germany
| | - Lars Pape
- Clinic for Paediatrics III, Essen University Hospital, Essen, Germany
| | - Lutz T. Weber
- Pediatric Nephrology, Children’s and Adolescents’ Hospital, University Hospital Cologne, Cologne, Germany
| | - Marcus Weitz
- University Hospital Tübingen, Pediatric Nephrology, Tübingen, Germany
| | - Jens König
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| | - Kai Krupka
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge and the NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| |
Collapse
|
37
|
de Marco R, Requião-Moura LR, Raimundo TRF, Mourão TB, Rampim GF, Medina-Pestana JO, Tedesco-Silva H, Gerbase-DeLima M. HLA-DPB1 molecular mismatches are risk factors for acute rejection and low 5-year graft function in first kidney transplants. HLA 2023; 101:228-238. [PMID: 36461794 DOI: 10.1111/tan.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/14/2022] [Accepted: 11/30/2022] [Indexed: 12/04/2022]
Abstract
The study aimed to investigate the impact of HLA-DPB1 allelic and molecular mismatches on the occurrence of acute rejection (AR) and low 5-year graft function (5Y-GF) in first kidney transplant (KT) recipients. This is a single center retrospective study of 130 deceased donor KT recipients transplanted between 2014 and 2016. HLA-DPB1 allelic MM and the following molecular MM (mMM) were analyzed: expression MM with the high expression G allele in the donor; T cell epitope MM (TCE MM); epitope MM (EMM), considering all six hypervariable regions (EMM-ABCDEF HVR), or only ABEF regions (EMM-ABEF HVR); eplet MM (EpMM); antibody-verified eplet MM (AbVer EpMM); and solvent accessible amino acid MM (SAMM). There was no association of allelic MM with AR or 5Y-GF. The variables independently associated (Cox regression analyses) with AR were high donor final creatinine, nonpermissive TCE MM, ABCDEF EMM load ≥6, EpMM load ≥6; SAMM load ≥5, and AbVer EpMM load ≥3. No association between any HLA-DPB1 mMM and 5Y-GF was observed when all 130 transplant recipients were considered. However, when transplants from expanded criteria donors were excluded, independent associations were detected (logistic regression analyses) with AbVerEpMM load ≥2, SAMM load ≥7, cerebro-vascular death, donor age, and AR. To our knowledge, this is the first study that shows that some HLA-DPB1 mMM are associated with AR and low 5Y-GF in a population of exclusively first kidney transplant recipients.
Collapse
Affiliation(s)
- Renato de Marco
- Instituto de Imunogenética (IGEN), Associação Fundo de Incentivo à Pesquisa (AFIP), São Paulo, Brazil
| | - Lúcio R Requião-Moura
- Nephrology Division, Hospital do Rim, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tamiris R F Raimundo
- Instituto de Imunogenética (IGEN), Associação Fundo de Incentivo à Pesquisa (AFIP), São Paulo, Brazil
| | - Tuíla B Mourão
- Instituto de Imunogenética (IGEN), Associação Fundo de Incentivo à Pesquisa (AFIP), São Paulo, Brazil
| | - Gisele F Rampim
- Instituto de Imunogenética (IGEN), Associação Fundo de Incentivo à Pesquisa (AFIP), São Paulo, Brazil
| | - José O Medina-Pestana
- Nephrology Division, Hospital do Rim, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Hélio Tedesco-Silva
- Nephrology Division, Hospital do Rim, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Gerbase-DeLima
- Instituto de Imunogenética (IGEN), Associação Fundo de Incentivo à Pesquisa (AFIP), São Paulo, Brazil
| |
Collapse
|
38
|
Knatterud ME, Simmons RL, Payne W, Stock P, Chavers B, Ascher N, Kaufman D, Kirk A, Keshavjee S, Humar A, Ganesh S, Hughes C, Kandaswamy R, Matas AJ. The John S. Najarian symposium: The past, present, and future of surgery and transplantation, May 20, 2022, Minneapolis, MN. Clin Transplant 2023; 37:e14877. [PMID: 36528870 DOI: 10.1111/ctr.14877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
Dr John S Najarian (1927-2020), chairman of the Department of Surgery at the University of Minnesota from 1967 to 1993, was a pioneer in surgery, clinical immunology and transplantation. A Covid-delayed Festschrift was held in his honor on May 20, 2022. The speakers reflected on his myriad contributions to surgery, transplantation, and resident/fellow training, as well as current areas of ongoing research to improve clinical outcomes. Of note, Dr Najarian was a founder of the journal Clinical Transplantation.
Collapse
Affiliation(s)
- Mary E Knatterud
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Richard L Simmons
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - William Payne
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Peter Stock
- Department of Surgery, University of California - San Francisco, San Francisco, California, USA
| | - Blanche Chavers
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Nancy Ascher
- Department of Surgery, University of California - San Francisco, San Francisco, California, USA
| | - Dixon Kaufman
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Alan Kirk
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Abhinav Humar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Swaytha Ganesh
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Christopher Hughes
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Raja Kandaswamy
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Arthur J Matas
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
39
|
Tambur AR, Das R. Can We Use Eplets (or Molecular) Mismatch Load Analysis to Improve Organ Allocation? The Hope and the Hype. Transplantation 2023; 107:605-615. [PMID: 36163639 PMCID: PMC9944744 DOI: 10.1097/tp.0000000000004307] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/14/2022] [Accepted: 07/03/2022] [Indexed: 11/25/2022]
Abstract
In recent years, there have been calls for implementation of "epitope matching" in deceased-donor organ allocation policies (later changed to "eplet matching"). Emerging data indeed support the use of molecular mismatch load analysis in specific patient groups, with the objective of posttransplant stratification into different treatment arms. For this purpose, the expectation is to statistically categorize patients as low- or high-immune-risk. Importantly, these patients will continue to be monitored' and their risk category, as well as their management, can be adjusted according to on-going findings. However, when discussing deceased donor organ allocation and matching algorithms, where the decision is not modifiable and has lasting impact on outcomes, the situation is fundamentally different. The goal of changing allocation schemes is to achieve the best possible HLA compatibility between donor and recipient. Immunologically speaking, this is a very different objective. For this purpose, the specific interplay of immunogenicity between the donor and any potential recipient must be understood. In seeking compatibility, the aim is not to redefine matching but to identify those mismatches that are "permissible" or' in other words, less immunogenic. In our eagerness to improve transplant outcome, unfortunately, we have conflated the hype with the hope. Terminology is used improperly, and new terms are created in the process with no sufficient support. Here, we call for a cautious evaluation of baseline assumptions and a critical review of the evidence to minimize unintended consequences.
Collapse
Affiliation(s)
- Anat R. Tambur
- Comprehensive Transplant Center, Department of Surgery, Northwestern University, Chicago, IL
| | - Rajdeep Das
- Comprehensive Transplant Center, Department of Surgery, Northwestern University, Chicago, IL
| |
Collapse
|
40
|
Bezstarosti S, Meziyerh S, Reinders MEJ, Voogt-Bakker K, Groeneweg KE, Roelen DL, Kers J, de Fijter JW, Heidt S. HLA-DQ eplet mismatch load may identify kidney transplant patients eligible for tacrolimus withdrawal without donor-specific antibody formation after mesenchymal stromal cell therapy. HLA 2023. [PMID: 36841928 DOI: 10.1111/tan.15008] [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: 10/17/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Recently, the randomized phase-II Triton study demonstrated that mesenchymal stromal cell (MSC) therapy facilitated early tacrolimus withdrawal in living donor kidney transplant recipients. The current sub-study analyzed formation of de novo donor-specific HLA antibodies (dnDSA) in the context of the degree of HLA eplet mismatches. At the time of protocol biopsy at 6 months, 7/29 patients (24%) in the MSC group and 1/27 patient (3.7%) in the control group had developed dnDSA. In the MSC group, all dnDSA were anti-HLA-DQ; two patients had anti-DQ alone and five patients combined with anti-class I, HLA-DR or -DP. Despite excess dnDSA formation in the MSC-arm of the study, the evolution of eGFR (CKD-EPI) and proteinuria were comparable 2 years posttransplant. All dnDSA were complement-binding and three patients had antibody-mediated rejection in the protocol biopsy, but overall rejection episodes were not increased. Everolimus had to be discontinued in nine patients because of toxicity, and tacrolimus was reintroduced in six patients because of dnDSA formation. The HLA-DQ eplet mismatch load independently associated with dnDSA (adjusted hazard ratio = 1.07 per eplet mismatch, p = 0.008). A threshold of ≥11 HLA-DQ eplet mismatches predicted subsequent dnDSA in all 11 patients in the MSC group, but specificity was low (44%). Further research is warranted to explore HLA molecular mismatch load as a biomarker to guide personalized maintenance immunosuppression in kidney transplantation.
Collapse
Affiliation(s)
- Suzanne Bezstarosti
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands.,Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Soufian Meziyerh
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Kim Voogt-Bakker
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Koen E Groeneweg
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesper Kers
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Johan W de Fijter
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands.,Eurotransplant Reference Laboratory, Leiden, The Netherlands
| |
Collapse
|
41
|
Seeking Standardized Definitions for HLA-incompatible Kidney Transplants: A Systematic Review. Transplantation 2023; 107:231-253. [PMID: 35915547 DOI: 10.1097/tp.0000000000004262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is no standard definition for "HLA incompatible" transplants. For the first time, we systematically assessed how HLA incompatibility was defined in contemporary peer-reviewed publications and its prognostic implication to transplant outcomes. METHODS We combined 2 independent searches of MEDLINE, EMBASE, and the Cochrane Library from 2015 to 2019. Content-expert reviewers screened for original research on outcomes of HLA-incompatible transplants (defined as allele or molecular mismatch and solid-phase or cell-based assays). We ascertained the completeness of reporting on a predefined set of variables assessing HLA incompatibility, therapies, and outcomes. Given significant heterogeneity, we conducted narrative synthesis and assessed risk of bias in studies examining the association between death-censored graft failure and HLA incompatibility. RESULTS Of 6656 screened articles, 163 evaluated transplant outcomes by HLA incompatibility. Most articles reported on cytotoxic/flow T-cell crossmatches (n = 98). Molecular genotypes were reported for selected loci at the allele-group level. Sixteen articles reported on epitope compatibility. Pretransplant donor-specific HLA antibodies were often considered (n = 143); yet there was heterogeneity in sample handling, assay procedure, and incomplete reporting on donor-specific HLA antibodies assignment. Induction (n = 129) and maintenance immunosuppression (n = 140) were frequently mentioned but less so rejection treatment (n = 72) and desensitization (n = 70). Studies assessing death-censored graft failure risk by HLA incompatibility were vulnerable to bias in the participant, predictor, and analysis domains. CONCLUSIONS Optimization of transplant outcomes and personalized care depends on accurate HLA compatibility assessment. Reporting on a standard set of variables will help assess generalizability of research, allow knowledge synthesis, and facilitate international collaboration in clinical trials.
Collapse
|
42
|
Tambur AR, Bestard O, Campbell P, Chong AS, Barrio MC, Ford ML, Gebel HM, Heidt S, Hickey M, Jackson A, Kosmoliaptsis V, Lefaucheur C, Louis K, Mannon RB, Mengel M, Morris A, Pinelli DF, Reed EF, Schinstock C, Taupin JL, Valenzuela N, Wiebe C, Nickerson P. Sensitization in transplantation: Assessment of Risk 2022 Working Group Meeting Report. Am J Transplant 2023; 23:133-149. [PMID: 36695615 DOI: 10.1016/j.ajt.2022.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/20/2022] [Accepted: 11/02/2022] [Indexed: 01/13/2023]
Abstract
The Sensitization in Transplantation: Assessment of Risk workgroup is a collaborative effort of the American Society of Transplantation and the American Society of Histocompatibility and Immunogenetics that aims at providing recommendations for clinical testing, highlights gaps in current knowledge, and proposes areas for further research to enhance histocompatibility testing in support of solid organ transplantation. This report provides updates on topics discussed by the previous Sensitization in Transplantation: Assessment of Risk working groups and introduces 2 areas of exploration: non-human leukocyte antigen antibodies and utilization of human leukocyte antigen antibody testing measurement to evaluate the efficacy of antibody-removal therapies.
Collapse
Affiliation(s)
- Anat R Tambur
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA.
| | - Oriol Bestard
- Vall d'Hebron Institut de Recerca, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Patricia Campbell
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Anita S Chong
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Martha Crespo Barrio
- Department of Nephrology, Hospital del Mar & Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, Georgia, USA
| | - Howard M Gebel
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Netherlands
| | - Michelle Hickey
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Annette Jackson
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| | - Kevin Louis
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| | - Roslyn B Mannon
- Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Michael Mengel
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Canada
| | - Anna Morris
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David F Pinelli
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | - Jean-Luc Taupin
- Department of Immunology, Saint Louis Hospital and University Paris-Cité, Paris, France
| | - Nicole Valenzuela
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter Nickerson
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
43
|
Senev A, Van Loon E, Lerut E, Coemans M, Callemeyn J, Daniëls L, Kerkhofs J, Koshy P, Kuypers D, Lamarthée B, Sprangers B, Tinel C, Van Craenenbroeck AH, Van Sandt V, Emonds MP, Naesens M. Association of Predicted HLA T-Cell Epitope Targets and T-Cell-Mediated Rejection After Kidney Transplantation. Am J Kidney Dis 2022; 80:718-729.e1. [PMID: 35690154 DOI: 10.1053/j.ajkd.2022.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 04/09/2022] [Indexed: 02/02/2023]
Abstract
RATIONALE & OBJECTIVE The relationship between human leukocyte antigen (HLA) molecular mismatches and T-cell-mediated rejection (TCMR) is unknown. We investigated the associations between the different donor HLA-derived T-cell targets and the occurrence of TCMR and borderline histologic changes suggestive of TCMR after kidney transplantation. STUDY DESIGN Retrospective cohort study. SETTING & PARTICIPANTS All kidney transplant recipients at a single center between 2004 and 2013 with available biopsy data and a DNA sample for high-resolution HLA donor/recipient typing (N = 893). EXPOSURE Scores calculated by the HLA matching algorithm PIRCHE-II and HLA eplet mismatches. OUTCOME TCMR, borderline changes suggestive of TCMR, and allograft failure. ANALYTICAL APPROACH Multivariable cause-specific hazards models were fit to characterize the association between HLA epitopes targets and study outcomes. RESULTS We found 277 patients developed TCMR, and 134 developed only borderline changes suggestive of TCMR on at least 1 biopsy. In multivariable analyses, only the PIRCHE-II scores for HLA-DRB1 and HLA-DQB1 were independently associated with the occurrence of TCMR and with allograft failure; this was not the case for HLA class I molecules. If restricted to rejection episodes within the first 3 months after transplantation, only the T-cell epitope targets originating from the donor's HLA-DRB1 and HLA-DQB1, but not class I molecules, were associated with the early acute TCMR. Also, the median PIRCHE-II score for HLA class II was statistically different between the patients with TCMR compared to the patients without TCMR (129 [IQR, 60-240] vs 201 [IQR, 96-298], respectively; P < 0.0001). These differences were not observed for class I PIRCHE-II scores. LIMITATIONS Observational clinical data and residual confounding. CONCLUSIONS In the absence of HLA-DSA, HLA class II but not class I mismatches are associated with early episodes of acute TCMR and allograft failure. This suggests that current immunosuppressive therapies are largely able to abort the most deleterious HLA class I-directed alloimmune processes; however, alloresponses against HLA-DRB1 and HLA-DQB1 molecular mismatches remain insufficiently suppressed. PLAIN-LANGUAGE SUMMARY Genetic differences in the human leukocyte antigen (HLA) complex between kidney transplant donors and recipients play a central role in T-cell-mediated rejection (TCMR), which can lead to failure of the transplanted kidney. Evaluating this genetic disparity (mismatch) in the HLA complex at the molecular (epitope) level could contribute to better prediction of the immune response to the donor organ posttransplantation. We investigated the associations of the different donor HLA-derived T-cell epitope targets and scores obtained from virtual crossmatch algorithms with the occurrence of TCMR, borderline TCMR, and graft failure after kidney transplantation after taking into account the influence of donor-specific anti-HLA antibodies. This study illustrates the greater importance of the molecular mismatches in class II molecules compared to class I HLA molecules.
Collapse
Affiliation(s)
- Aleksandar Senev
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium; Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Elisabet Van Loon
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium
| | - Evelyne Lerut
- Department of Imaging & Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Coemans
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium
| | - Jasper Callemeyn
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium
| | - Liesbeth Daniëls
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Johan Kerkhofs
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Priyanka Koshy
- Department of Imaging & Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Kuypers
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Baptiste Lamarthée
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium
| | - Ben Sprangers
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Claire Tinel
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium
| | - Amaryllis H Van Craenenbroeck
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Vicky Van Sandt
- Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Marie-Paule Emonds
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium; Histocompatibility and Immunogenetics Laboratory (HILA), Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Maarten Naesens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, KU Leuven University, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
44
|
Chancay J, Liu C, Chauhan K, Andersen L, Harris C, Coca S, Delaney V, Tedla F, De Boccardo G, Sehgal V, Moledina D, Formica R, Reghuvaran A, Banu K, Florman S, Akalin E, Shapiro R, Salem F, Menon MC. Role of time from transplantation to biopsy in histologic ABMR: A single center report. Clin Transplant 2022; 36:e14802. [PMID: 36069577 PMCID: PMC10211409 DOI: 10.1111/ctr.14802] [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: 01/15/2022] [Revised: 06/24/2022] [Accepted: 08/19/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Allograft biopsies with lesions of Antibody-Mediated Rejection (ABMR) with Microvascular Inflammation (MVI) have shown heterogeneous etiologies and outcomes. METHODS To examine factors associated with outcomes in biopsies that meet histologic ABMR criteria, we retrospectively evaluated for-cause biopsies at our center between 2011 and 2017. We included biopsies that met the diagnosis of ABMR by histology, along with simultaneous evaluation for anti-Human Leukocyte Antigen (HLA) donor-specific antibodies (DSA). We evaluated death-censored graft loss (DCGL) and used a principal component analysis (PCA) approach to identify key predictors of outcomes. RESULTS Out of the histologic ABMR cohort (n = 118), 70 were DSA-positive ABMR, while 48 had no DSA. DSA(+)ABMR were younger and more often female recipients. DSA(+)ABMR occurred significantly later post-transplant than DSA(-)ABMR suggesting time-dependence. DSA(+)ABMR had higher inflammatory scores (i,t), chronicity scores (ci, ct) and tended to have higher MVI scores. Immunodominance of DQ-DSA in DSA(+)ABMR was associated with higher i+t scores. Clinical/histologic factors significantly associated with DCGL after biopsy were inputted into the PCA. Principal component-1 (PC-1), which contributed 34.8% of the variance, significantly correlated with time from transplantation to biopsy, ci/ct scores and DCGL. In the PCA analyses, i, t scores, DQ-DSA, and creatinine at biopsy retained significant correlations with GL-associated PCs. CONCLUSIONS Time from transplantation to biopsy plays a major role in the prognosis of biopsies with histologic ABMR and MVI, likely due to ongoing chronic allograft injury over time.
Collapse
Affiliation(s)
- Jorge Chancay
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Caroline Liu
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kinsuk Chauhan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lisa Andersen
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cynthia Harris
- Transplant Center at Massachusetts General Hospital, Harvard Medical School, Cambridge, MA, United States
| | - Steven Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Veronica Delaney
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fasika Tedla
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Graciela De Boccardo
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Vinita Sehgal
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dennis Moledina
- Yale New Haven Transplantation Center, Yale School of Medicine, New Haven, CT, United States
| | - Richard Formica
- Yale New Haven Transplantation Center, Yale School of Medicine, New Haven, CT, United States
| | - Anand Reghuvaran
- Yale New Haven Transplantation Center, Yale School of Medicine, New Haven, CT, United States
| | - Khadija Banu
- Yale New Haven Transplantation Center, Yale School of Medicine, New Haven, CT, United States
| | - Sander Florman
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Enver Akalin
- Montefiore Einstein Center for Transplantation, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ron Shapiro
- Transplant Center at Massachusetts General Hospital, Harvard Medical School, Cambridge, MA, United States
| | - Fadi Salem
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Madhav C Menon
- Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Yale New Haven Transplantation Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
45
|
Nickerson PW. Rationale for the IMAGINE study for chronic active antibody-mediated rejection (caAMR) in kidney transplantation. Am J Transplant 2022; 22 Suppl 4:38-44. [PMID: 36453707 DOI: 10.1111/ajt.17210] [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: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Chronic active antibody-mediated rejection (caAMR) in kidney transplantation is a major cause of late graft loss and despite all efforts to date, there is no proven effective therapy. Indeed, the Transplant Society (TTS) consensus opinion called for a conservative approach optimizing baseline immunosuppression and supportive care focused on blood pressure, blood glucose, and lipid control. This review provides the rationale and early evidence in kidney transplant recipients with caAMR that supported the design of the IMAGINE study whose goal is to evaluate the potential impact of targeting the IL6/IL6R pathway.
Collapse
Affiliation(s)
- Peter W Nickerson
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.,Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
46
|
Wiebe C, Nickerson PW, Kosmoliaptsis V. Molecular Mismatch and the Risk for T Cell-Mediated Rejection. Am J Kidney Dis 2022; 80:704-706. [PMID: 36057468 DOI: 10.1053/j.ajkd.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 02/02/2023]
Affiliation(s)
- Chris Wiebe
- Department of Medicine, University of Manitoba, Winnipeg, Canada; Shared Health Services Manitoba; Department of Immunology, University of Manitoba, Winnipeg, Canada.
| | - Peter W Nickerson
- Department of Medicine, University of Manitoba, Winnipeg, Canada; Shared Health Services Manitoba; Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom, and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge; NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| |
Collapse
|
47
|
Masset C, Dantal J, Soulillou JP, Walencik A, Delbos F, Brouard S, Giral M. Case Report: Long-term observations from the tacrolimus weaning randomized clinical trial depicts the challenging aspects for determination of low-immunological risk patients. Front Immunol 2022; 13:1021481. [PMID: 36518770 PMCID: PMC9744190 DOI: 10.3389/fimmu.2022.1021481] [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: 08/17/2022] [Accepted: 10/31/2022] [Indexed: 11/30/2022] Open
Abstract
Whilst calcineurin inhibitors (CNI) are the cornerstone of immunosuppressive maintenance therapy in kidney transplantation, several studies have investigated the safety of CNI withdrawal in order to avoid their numerous side effects. In this context, we performed several years ago a clinical randomized trial evaluating CNI weaning in stable kidney transplant recipients without anti-HLA immunization. The trial was interrupted prematurely due to a high number of de novo DSA (dnDSA) and biopsy proven acute rejection (BPAR) in patients who underwent tacrolimus weaning, resulting in treatment for rejection and resumption of tacrolimus. We report here the long-term outcomes of patients included in this clinical trial. Ten years after randomization, all patients are alive with a functional allograft. They all receive tacrolimus therapy except one with recurrent cutaneous neoplasia issues. Long-term eGFR was comparable between patients of the two randomized groups (46.4 ml/min vs 42.8 ml/min). All dnDSA that occurred during the study period became non-detectable and all rejections episodes were reversed. The retrospective assessment of HLA DQ single molecule epitope mismatching determined that a majority of patients who developed dnDSA after tacrolimus withdrawal would have been considered at high immunological risk. Minimization of immunosuppression remains a challenging objective, mainly because of the issues to properly select very low immunological risk patients. Valuable improvements have been made the last decade regarding evaluation of the allograft rejection notably through the determination of numerous at-risk biomarkers. However, even if the impact of such tools still need to be clarify in clinical routine, they may permit an improvement in patients' selection for immunosuppression minimization without increasing the risk of allograft rejection.
Collapse
Affiliation(s)
- Christophe Masset
- Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalo-Universitaire (CHU) Nantes, Nantes, France,Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jacques Dantal
- Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalo-Universitaire (CHU) Nantes, Nantes, France,Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jean-Paul Soulillou
- Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalo-Universitaire (CHU) Nantes, Nantes, France,Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Alexandre Walencik
- Laboratoire d’immunologie et HLA Etablissement Français du Sang, Nantes, France
| | - Florent Delbos
- Laboratoire d’immunologie et HLA Etablissement Français du Sang, Nantes, France
| | - Sophie Brouard
- Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalo-Universitaire (CHU) Nantes, Nantes, France,Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France,*Correspondence: Magali Giral, ; Sophie Brouard,
| | - Magali Giral
- Institut de Transplantation Urologie Néphrologie (ITUN), Centre Hospitalo-Universitaire (CHU) Nantes, Nantes, France,Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France,*Correspondence: Magali Giral, ; Sophie Brouard,
| | | |
Collapse
|
48
|
Lemieux W, Fleischer D, Yang AY, Niemann M, Oualkacha K, Klement W, Richard L, Polychronakos C, Liwski R, Claas F, Gebel HM, Keown PA, Lewin A, Sapir-Pichhadze R. Dissecting the impact of molecular T-cell HLA mismatches in kidney transplant failure: A retrospective cohort study. Front Immunol 2022; 13:1067075. [PMID: 36505483 PMCID: PMC9730505 DOI: 10.3389/fimmu.2022.1067075] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Kidney transplantation is the optimal treatment in end-stage kidney disease, but de-novo donor specific antibody development continues to negatively impact patients undergoing kidney transplantation. One of the recent advances in solid organ transplantation has been the definition of molecular mismatching between donors and recipients' Human Leukocyte Antigens (HLA). While not fully integrated in standard clinical care, cumulative molecular mismatch at the level of eplets (EMM) as well as the PIRCHE-II score have shown promise in predicting transplant outcomes. In this manuscript, we sought to study whether certain T-cell molecular mismatches (TcEMM) were highly predictive of death-censored graft failure (DCGF). Methods We studied a retrospective cohort of kidney donor:recipient pairs from the Scientific Registry of Transplant Recipients (2000-2015). Allele level HLA-A, B, C, DRB1 and DQB1 types were imputed from serologic types using the NMDP algorithm. TcEMMs were then estimated using the PIRCHE-II algorithm. Multivariable Accelerated Failure Time (AFT) models assessed the association between each TcEMM and DCGF. To discriminate between TcEMMs most predictive of DCGF, we fit multivariable Lasso penalized regression models. We identified co-expressed TcEMMs using weighted correlation network analysis (WGCNA). Finally, we conducted sensitivity analyses to address PIRCHE and IMGT/HLA version updates. Results A total of 118,309 donor:recipient pairs meeting the eligibility criteria were studied. When applying the PIRCHE-II algorithm, we identified 1,935 distinct TcEMMs at the population level. A total of 218 of the observed TcEMM were independently associated with DCGF by AFT models. The Lasso penalized regression model with post selection inference identified a smaller subset of 86 TcEMMs (56 and 30 TcEMM derived from HLA Class I and II, respectively) to be highly predictive of DCGF. Of the observed TcEMM, 38.14% appeared as profiles of highly co-expressed TcEMMs. In addition, sensitivity analyses identified that the selected TcEMM were congruent across IMGT/HLA versions. Conclusion In this study, we identified subsets of TcEMMs highly predictive of DCGF and profiles of co-expressed mismatches. Experimental verification of these TcEMMs determining immune responses and how they may interact with EMM as predictors of transplant outcomes would justify their consideration in organ allocation schemes and for modifying immunosuppression regimens.
Collapse
Affiliation(s)
- William Lemieux
- Centre for Outcomes Research and Evaluation (CORE), Research Institute of McGill University Health Centre, Montréal, QC, Canada,Medical Affairs & Innovation, Héma-Québec, Montréal, QC, Canada
| | - David Fleischer
- Department of Mathematics and Statistics, McGill University, Montreal, QC, Canada
| | - Archer Yi Yang
- Department of Mathematics and Statistics, McGill University, Montreal, QC, Canada
| | | | - Karim Oualkacha
- Department of Mathematics, Université du Québec à Montreal, Montreal, QC, Canada
| | - William Klement
- Division of Organ Donation and Transplantation, Canadian Blood Services, Ottawa, ON, Canada
| | - Lucie Richard
- Transfusion medicine/Reference Laboratory, Héma-Québec, Montréal, QC, Canada
| | - Constantin Polychronakos
- Department of Pediatrics, The Research Institute of the McGill University Health Centre and the Montreal Children’s Hospital, Montréal, QC, Canada
| | - Robert Liwski
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Frans Claas
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Howard M. Gebel
- Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| | - Paul A. Keown
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Antoine Lewin
- Medical Affairs & Innovation, Héma-Québec, Montréal, QC, Canada,Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Ruth Sapir-Pichhadze
- Centre for Outcomes Research and Evaluation (CORE), Research Institute of McGill University Health Centre, Montréal, QC, Canada,Division of Nephrology and the Multi-Organ Transplant Program, Royal Victoria Hospital, McGill University Health Centre, Montréal, QC, Canada,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC, Canada,*Correspondence: Ruth Sapir-Pichhadze,
| |
Collapse
|
49
|
Parajuli S, Hidalgo LG, Foley D. Immunology of simultaneous liver and kidney transplants with identification and prevention of rejection. FRONTIERS IN TRANSPLANTATION 2022; 1:991546. [PMID: 38994375 PMCID: PMC11235231 DOI: 10.3389/frtra.2022.991546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/12/2022] [Indexed: 07/13/2024]
Abstract
Simultaneous liver and kidney (SLK) transplantation is considered the best treatment modality among selected patients with both chronic kidney disease (CKD) and end-stage liver disease (ESLD). Since the first SLK transplant in 1983, the number of SLK transplants has increased worldwide, and particularly in the United States since the implementation of the MELD system in 2002. SLK transplants are considered a relatively low immunological risk procedure evidenced by multiple studies displaying the immunomodulatory properties of the liver on the immune system of SLK recipients. SLK recipients demonstrate lower rates of both cellular and antibody-mediated rejection on the kidney allograft when compared to kidney transplant-alone recipients. Therefore, SLK transplants in the setting of preformed donor-specific HLA antibodies (DSA) are a common practice, at many centers. Acceptance and transplantation of SLKs are based solely on ABO compatibility without much consideration of crossmatch results or DSA levels. However, some studies suggest an increased risk for rejection for SLK recipients transplanted across high levels of pre-formed HLA DSA. Despite this, there is no consensus regarding acceptable levels of pre-formed DSA, the role of pre-transplant desensitization, splenectomy, or immunosuppressive management in this unique population. Also, the impact of post-transplant DSA monitoring on long-term outcomes is not well-studied in SLK recipients. In this article, we review recent and relevant past articles in this field with a focus on the immunological risk factors among SLK recipients, and strategies to mitigate the negative outcomes among them.
Collapse
Affiliation(s)
- Sandesh Parajuli
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Luis G Hidalgo
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - David Foley
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
50
|
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: 3.5] [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
|