1
|
Lombardi Y, Raynaud M, Schatzl M, Mayer KA, Diebold M, Patel UD, Schrezenmeier E, Akifova A, Budde K, Loupy A, Böhmig GA. Estimating the efficacy of felzartamab to treat antibody-mediated rejection using the iBox prognostication system. Am J Transplant 2025; 25:1130-1132. [PMID: 39674514 DOI: 10.1016/j.ajt.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/26/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Affiliation(s)
- Yannis Lombardi
- Paris Transplant Group, Paris Institute for Transplantation & Organ Regeneration, Inserm/Université Paris Cité, Paris, France; Department of Nephrology, Tenon Hospital, Assistance Publique-Hôpitaux de Paris/Sorbonne University, Paris, France
| | - Marc Raynaud
- Paris Transplant Group, Paris Institute for Transplantation & Organ Regeneration, Inserm/Université Paris Cité, Paris, France
| | - Martina Schatzl
- Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Katharina A Mayer
- Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matthias Diebold
- Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Uptal D Patel
- Human Immunology Biosciences, Inc (HI-Bio), South San Francisco, California, USA
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Aylin Akifova
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandre Loupy
- Paris Transplant Group, Paris Institute for Transplantation & Organ Regeneration, Inserm/Université Paris Cité, Paris, France.
| | - Georg A Böhmig
- Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Truchot A, Raynaud M, Helanterä I, Aubert O, Kamar N, Divard G, Astor B, Legendre C, Hertig A, Buchler M, Crespo M, Akalin E, Pujol GS, Ribeiro de Castro MC, Matas AJ, Ulloa C, Jordan SC, Huang E, Juric I, Basic-Jukic N, Coemans M, Naesens M, Friedewald JJ, Silva HT, Lefaucheur C, Segev DL, Collins GS, Loupy A. Competing and Noncompeting Risk Models for Predicting Kidney Allograft Failure. J Am Soc Nephrol 2025; 36:688-701. [PMID: 40168162 PMCID: PMC11975249 DOI: 10.1681/asn.0000000517] [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: 05/31/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Background Prognostic models are becoming increasingly relevant in clinical trials as potential surrogate end points and for patient management as clinical decision support tools. However, the effect of competing risks on model performance remains poorly investigated. We aimed to carefully assess the performance of competing risk and noncompeting risk models in the context of kidney transplantation, where allograft failure and death with a functioning graft are two competing outcomes. Methods We included 11,046 kidney transplant recipients enrolled in ten countries. We developed prediction models for long-term kidney graft failure prediction, without accounting (i.e., censoring) and accounting for the competing risk of death with a functioning graft, using Cox, Fine–Gray, and cause-specific Cox regression models. To this aim, we followed a detailed and transparent analytical framework for competing and noncompeting risk modeling and carefully assessed the models' development, stability, discrimination, calibration, overall fit, clinical utility, and generalizability in external validation cohorts and subpopulations. More than 15 metrics were used to provide an exhaustive assessment of model performance. Results Among 11,046 recipients in the derivation and validation cohorts, 1497 (14%) lost their graft and 1003 (9%) died with a functioning graft after a median follow-up postrisk evaluation of 4.7 years (interquartile range, 2.7–7.0). The cumulative incidence of graft loss was similarly estimated by Kaplan–Meier and Aalen–Johansen methods (17% versus 16% in the derivation cohort). Cox and competing risk models showed similar and stable risk estimates for predicting long-term graft failure (average mean absolute prediction error of 0.0140, 0.0138, and 0.0135 for Cox, Fine–Gray, and cause-specific Cox models, respectively). Discrimination and overall fit were comparable in the validation cohorts, with concordance index ranging from 0.76 to 0.87. Across various subpopulations and clinical scenarios, the models performed well and similarly, although in some high-risk groups (such as donors older than 65 years), the findings suggest a trend toward moderately improved calibration when using a competing risk approach. Conclusions Competing and noncompeting risk models performed similarly in predicting long-term kidney graft failure.
Collapse
Affiliation(s)
- Agathe Truchot
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Marc Raynaud
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
| | - Ilkka Helanterä
- Department of Transplantation and Liver Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Olivier Aubert
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, Toulouse Rangueil University Hospital, INSERM UMR 1291, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University Paul Sabatier, Toulouse, France
| | - Gillian Divard
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Brad Astor
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Christophe Legendre
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Alexandre Hertig
- Department of Nephrology and Kidney Transplantation, Foch Hospital, Suresnes, France
| | - Matthias Buchler
- Nephrology and Immunology Department, Bretonneau Hospital, Tours, France
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar Barcelona, Barcelona, Spain
| | - Enver Akalin
- Kidney Transplantation Program, Renal Division Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Gervasio Soler Pujol
- Centro de Educacion Medica e Investigaciones Clinicas Buenos Aires, Unidad de Trasplante Renopancreas, Buenos Aires, Argentina
| | | | - Arthur J. Matas
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | | | - Stanley C. Jordan
- Division of Nephrology, Department of Medicine, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Edmund Huang
- Division of Nephrology, Department of Medicine, Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Ivana Juric
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Nikolina Basic-Jukic
- Department of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | | | - Helio Tedesco Silva
- Hospital do Rim, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Dorry L. Segev
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gary S. Collins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Centre for Statistics in Medicine, University of Oxford, Oxford, United Kingdom
| | - Alexandre Loupy
- INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, Université de Paris Cité, Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France
| |
Collapse
|
3
|
Diebold M, Mayer KA, Hidalgo L, Kozakowski N, Budde K, Böhmig GA. Chronic Rejection After Kidney Transplantation. Transplantation 2025; 109:610-621. [PMID: 39192468 PMCID: PMC11927446 DOI: 10.1097/tp.0000000000005187] [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: 05/28/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [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
|
4
|
Walker H, McLeman L, Meyran D, Goh LY, Summers P, Stolper J, Hanna D, Hughes D, Wang S, Toro C, Williams E, Dyas R, Rubinek LC, Taylor K, Selman CJ, Grobler A, Lee KJ, Snelling T, Cole T, Gwee A, Conyers R. Co-designing a Novel Ordinal Endpoint for an Adaptive Platform Trial, BANDICOOT, in Pediatric Hematopoietic Stem Cell Transplant. Transplant Cell Ther 2025:S2666-6367(25)01007-3. [PMID: 39921207 DOI: 10.1016/j.jtct.2025.01.894] [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: 12/20/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
An adaptive platform trial (APT) offers the ability to incorporate several research questions in the same target population across multiple domains (interventions), with the ability to add new questions in a perpetual manner. An APT is particularly appealing for pediatric hematopoietic stem cell transplant (HCT); an area of high heterogeneity, limited trial availability, and high mortality. Ideally, all domains in an APT would have the same primary endpoint. Therefore, an ordinal endpoint with multiple categories that combines various clinical outcomes into a single outcome measure is particularly appealing for APTs. Unfortunately, there is no accepted ordinal endpoint for pediatric HCT trials. This article aims to describe the methodology used to co-design a novel primary ordinal endpoint for the pediatric HCT APT - BANDICOOT. BANDICOOT is a study that aims to build an adaptive novel platform design - improving the complications, cost-effectiveness, outcomes, and overall survival from hematopoietic stem cell transplantation.The results of this process identified two potential ordinal endpoints that could be used, one focusing on organ support and the other on a combination of organ support, viral reactivation, and immune reconstitution. We explored the data extraction required for these endpoints from electronic medical records that we will utilize to validate the endpoints and determine which will be used in the APT BANDICOOT. In an era in which APTs are becoming increasingly utilized to answer important questions in clinical care, this article describes a reproducible strategy for the design of high-quality and meaningful ordinal endpoints.
Collapse
Affiliation(s)
- Hannah Walker
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Lorna McLeman
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Deborah Meyran
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Li-Yin Goh
- Centre for Health Analytics, Murdoch Children's Research Institute, Parkville, Australia
| | - Peter Summers
- Centre for Health Analytics, Murdoch Children's Research Institute, Parkville, Australia
| | - Julian Stolper
- Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Diane Hanna
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - David Hughes
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia
| | - Stacie Wang
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia
| | - Claudia Toro
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Elizabeth Williams
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Roxanne Dyas
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Lori Chait Rubinek
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Kaitlyn Taylor
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia
| | - Chris J Selman
- Department of Paediatrics, University of Melbourne, Parkville, Australia; Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Australia
| | - Anneke Grobler
- Department of Paediatrics, University of Melbourne, Parkville, Australia; Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Australia
| | - Katherine J Lee
- Department of Paediatrics, University of Melbourne, Parkville, Australia; Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Australia
| | - Tom Snelling
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia
| | - Theresa Cole
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Amanda Gwee
- Department of Paediatrics, University of Melbourne, Parkville, Australia; Infectious Diseases Department, Royal Children's Hospital, Parkville, Australia; Antimicrobials Group, Murdoch Children's Research Institute, Parkville, Australia
| | - Rachel Conyers
- Children's Cancer Centre, Royal Children's Hospital, Parkville, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Australia.
| |
Collapse
|
5
|
Coemans M, Tran TH, Döhler B, Massie AB, Verbeke G, Segev DL, Gentry SE, Naesens M. A competing risks model to estimate the risk of graft failure and patient death after kidney transplantation using continuous donor-recipient age combinations. Am J Transplant 2025; 25:355-367. [PMID: 39111667 DOI: 10.1016/j.ajt.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 09/08/2024]
Abstract
Graft failure and recipient death with functioning graft are important competing outcomes after kidney transplantation. Risk prediction models typically censor for the competing outcome thereby overestimating the cumulative incidence. The magnitude of this overestimation is not well described in real-world transplant data. This retrospective cohort study analyzed data from the European Collaborative Transplant Study (n = 125 250) and from the American Scientific Registry of Transplant Recipients (n = 190 258). Separate cause-specific hazard models using donor and recipient age as continuous predictors were developed for graft failure and recipient death. The hazard of graft failure increased quadratically with increasing donor age and decreased decaying with increasing recipient age. The hazard of recipient death increased linearly with increasing donor and recipient age. The cumulative incidence overestimation due to competing risk-censoring was largest in high-risk populations for both outcomes (old donors/recipients), sometimes amounting to 8.4 and 18.8 percentage points for graft failure and recipient death, respectively. In our illustrative model for posttransplant risk prediction, the absolute risk of graft failure and death is overestimated when censoring for the competing event, mainly in older donors and recipients. Prediction models for absolute risks should treat graft failure and death as competing events.
Collapse
Affiliation(s)
- Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Public Health & Primary Care, Leuven Biostatistics and Statistical Bioinformatics Centre (L-Biostat), KU Leuven, Leuven, Belgium
| | - Thuong Hien Tran
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Bernd Döhler
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Allan B Massie
- Department of Surgery, NYU Grossman School of Medicine, New York, New York, USA; Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Geert Verbeke
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-Biostat), Universiteit Hasselt and KU Leuven, Hasselt and Leuven, Belgium
| | - Dorry L Segev
- Department of Surgery, NYU Grossman School of Medicine, New York, New York, USA; Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Sommer E Gentry
- Department of Surgery, NYU Grossman School of Medicine, New York, New York, USA; Department of Population Health, NYU Grossman School of Medicine, New York, New York, USA
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Savoye E, Santin G, Legeai C, Kerbaul F, Gaillard F, Pastural M. Comparison of Kidney Graft Function and Survival in an Emulated Trial With Living Donors and Brain-Dead Donors. Transpl Int 2024; 37:13208. [PMID: 39267619 PMCID: PMC11391114 DOI: 10.3389/ti.2024.13208] [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: 04/30/2024] [Accepted: 07/25/2024] [Indexed: 09/15/2024]
Abstract
Living donation (LD) transplantation is the preferred treatment for kidney failure as compared to donation after brain death (DBD), but age may play a role. We compared the 1-year estimated glomerular filtration rate (eGFR) after kidney transplantation for recipients of LD and DBD stratified by recipient and donor age between 2015 and 2018 in a matched cohort. The strength of the association between donation type and 1-year eGFR differed by recipient age (P interaction < 0.0001). For LD recipients aged 40-54 years versus same-aged DBD recipients, the adjusted odds ratio (aOR) for eGFR ≥60 mL/min/1.73 m2 was 1.48 (95% CI: 1.16-1.90). For DBD recipients aged ≥ 60 years, the aOR was 0.18 (95% CI: 0.12-0.29) versus DBD recipients aged 40-54 years but was 0.91 (95% CI: 0.67-1.24) versus LD recipients aged ≥60 years. In the matched cohort, 4-year graft and patient survival differed by donor age and type. As compared with DBD grafts, LD grafts increased the proportion of recipients with 1-year eGFR ≥60 mL/min/1.73 m2. Recipients aged ≥60 years benefited most from LD transplantation, even if the donor was aged ≥60 years. For younger recipients, large age differences between donor and recipient could also be addressed with a paired exchange program.
Collapse
Affiliation(s)
- Emilie Savoye
- Direction Prélèvement Greffe Organes-Tissus, Agence de la Biomédecine, Saint-Denis La Plaine, France
| | - Gaëlle Santin
- Direction Prélèvement Greffe Organes-Tissus, Agence de la Biomédecine, Saint-Denis La Plaine, France
| | - Camille Legeai
- Direction Prélèvement Greffe Organes-Tissus, Agence de la Biomédecine, Saint-Denis La Plaine, France
| | - François Kerbaul
- Direction Prélèvement Greffe Organes-Tissus, Agence de la Biomédecine, Saint-Denis La Plaine, France
| | - François Gaillard
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Myriam Pastural
- Direction Prélèvement Greffe Organes-Tissus, Agence de la Biomédecine, Saint-Denis La Plaine, France
| |
Collapse
|
7
|
Chauvelot L, Barba T, Saison C, Siska E, Kulifaj D, Bakker SJL, Koenig A, Rabeyrin M, Buron F, Picard C, Dijoud F, Manière L, Lina B, Morelon E, Dubois V, Thaunat O. Longitudinal monitoring of Torque Teno virus DNAemia in kidney transplant recipients correlates with long-term complications of inadequate immunosuppression. J Med Virol 2024; 96:e29806. [PMID: 39007420 DOI: 10.1002/jmv.29806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/20/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024]
Abstract
Optimization of individual immunosuppression, which reduces the risks of both graft loss and patients' death, is considered the best approach to improve long-term outcomes of renal transplantation. Torque Teno Virus (TTV) DNAemia has emerged as a potential biomarker reflecting the depth of therapeutic immunosuppression during the initial year post-transplantation. However, its efficacy in long-term monitoring remains uncertain. In a cohort study involving 34 stable kidney transplant recipients and 124 healthy volunteers, we established lower and upper TTV DNAemia thresholds (3.75-5.1 log10 cp/mL) correlating with T-cell activatability, antibody response against flu vaccine, and risk for subsequent serious infections or cancer over 50 months. Validation in an independent cohort of 92 recipients confirmed that maintaining TTV DNAemia within this range in >50% of follow-up time points was associated with reduced risks of complications due to inadequate immunosuppression, including de novo DSA, biopsy-proven antibody-mediated rejection, graft loss, infections, or cancer. Multivariate analysis highlighted "in-target" TTV DNAemia as the sole independent variable significantly linked to decreased risk for long-term complications due to inadequate immunosuppression (odds ratio [OR]: 0.27 [0.09-0.77]; p = 0.019). Our data suggest that the longitudinal monitoring of TTV DNAemia in kidney transplant recipients could help preventing the long-term complications due to inadequate immunosuppression.
Collapse
Affiliation(s)
- Luc Chauvelot
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Thomas Barba
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
- Department of Internal Medicine, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
| | - Carole Saison
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- French National Blood Service (EFS), HLA Laboratory, Lyon, France
| | - Evangelia Siska
- BioMérieux SA, 138, Rue Louis PASTEUR, Parc Technologique Delta Sud, Verniolle, France
| | - Dorian Kulifaj
- BioMérieux SA, 138, Rue Louis PASTEUR, Parc Technologique Delta Sud, Verniolle, France
| | - Stephan J L Bakker
- Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Alice Koenig
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, Bron, France
| | - Fanny Buron
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
| | - Cécile Picard
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, Bron, France
| | - Frédérique Dijoud
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, Bron, France
| | - Louis Manière
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
| | - Bruno Lina
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Emmanuel Morelon
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Valerie Dubois
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- French National Blood Service (EFS), HLA Laboratory, Lyon, France
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Groupement Hospitalier Centre, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
8
|
Balakrishnan S, Alexander MP, Schinstock C. Challenges and opportunities for designing clinical trials for antibody mediated rejection. FRONTIERS IN TRANSPLANTATION 2024; 3:1389005. [PMID: 38993760 PMCID: PMC11235363 DOI: 10.3389/frtra.2024.1389005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/25/2024] [Indexed: 07/13/2024]
Abstract
Significant progress has been made in kidney transplantation, with 1-year graft survival nearing 95%. However, long-term allograft survival remains suboptimal, with a 10-year overall graft survival rate of only 53.6% for deceased donor transplant recipients. Chronic active antibody-mediated rejection (ABMR) is a leading cause of death-censored graft loss, yet no therapy has demonstrated efficacy in large, randomized trials, despite substantial investment from pharmaceutical companies. Several clinical trials aimed to treat chronic ABMR in the past decade have yielded disappointing results or were prematurely terminated, attributed to factors including incomplete understanding of disease mechanisms, heterogeneous patient populations with comorbidities, slow disease progression, and limited patient numbers. This review aims to discuss opportunities for improving retrospective and prospective studies of ABMR, focusing on addressing heterogeneity, outcome measurement, and strategies to enhance patient enrollment to inform study design, data collection, and reporting.
Collapse
Affiliation(s)
- Suryanarayanan Balakrishnan
- Division of Hypertension and Nephrology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Mariam P. Alexander
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Carrie Schinstock
- Division of Hypertension and Nephrology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
9
|
Solez K, Eknoyan G. Transplant nephropathology: Wherefrom, wherein, and whereto. Clin Transplant 2024; 38:e15309. [PMID: 38619321 DOI: 10.1111/ctr.15309] [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: 06/13/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/16/2024]
Abstract
Renal pathology is a relatively recent entry in nephrology. While diseases of the kidney are old, their study began in the 19th century with the report of Richard Bright of the lesions of end-stage kidney disease. Its easy diagnosis from albuminuria soon elevated Bright's nephritis into a leading cause of death. The transformative events in the care of these cases were renal replacement therapy that converted a fatal into a chronic disease, and kidney biopsy that allowed study of the course and pathogenesis of kidney disease. Apart from its fundamental contributions to clinical nephrology, biopsy of renal allografts became an integral component of the evaluation and care of kidney transplant recipients. The Banff transplant pathology conferences launched in 1991 led to developing the classification of allograft pathology into an essential element in the evaluation, treatment, and care of allograft recipients with spirit of discovery. That success came at the cost of increasing complexity leading to the recent realization that it may need the refinement of its consensus-based system into a more evidence-based system with graded statements that are easily accessible to the other disciplines involved in the care of transplanted patients. Collaboration with other medical disciplines, allowing public comment on meeting reports, and incorporation of generative artificial intelligence (AI) are important elements of a successful future. The increased pace of innovation brought about by AI will likely allow us to solve the organ shortage soon and require new classifications for xenotransplantation pathology, tissue engineering pathology, and bioartificial organ pathology.
Collapse
Affiliation(s)
- Kim Solez
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Garabed Eknoyan
- The Selzman Institute of Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, USA
| |
Collapse
|
10
|
Streichart L, Felldin M, Ekberg J, Mjörnstedt L, Lindnér P, Lennerling A, Bröcker V, Mölne J, Holgersson J, Daenen K, Wennberg L, Lorant T, Baid-Agrawal S. Tocilizumab in chronic active antibody-mediated rejection: rationale and protocol of an in-progress randomized controlled open-label multi-center trial (INTERCEPT study). Trials 2024; 25:213. [PMID: 38519988 PMCID: PMC10958896 DOI: 10.1186/s13063-024-08020-0] [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: 12/05/2023] [Accepted: 02/26/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Chronic active antibody-mediated rejection (caAMR) in kidney transplants is associated with irreversible tissue damage and a leading cause of graft loss in the long-term. However, the treatment for caAMR remains a challenge to date. Recently, tocilizumab, a recombinant humanized monoclonal antibody directed against the human interleukin-6 (IL-6) receptor, has shown promise in the treatment of caAMR. However, it has not been systematically investigated so far underscoring the need for randomized controlled studies in this area. METHODS The INTERCEPT study is an investigator-driven randomized controlled open-label multi-center trial in kidney transplant recipients to assess the efficacy of tocilizumab in the treatment of biopsy-proven caAMR. A total of 50 recipients with biopsy-proven caAMR at least 12 months after transplantation will be randomized to receive either tocilizumab (n = 25) added to our standard of care (SOC) maintenance treatment or SOC alone (n = 25) for a period of 24 months. Patients will be followed for an additional 12 months after cessation of study medication. After the inclusion biopsies at baseline, protocol kidney graft biopsies will be performed at 12 and 24 months. The sample size calculation assumed a difference of 5 ml/year in slope of estimated glomerular filtration rate (eGFR) between the two groups for 80% power at an alpha of 0.05. The primary endpoint is the slope of eGFR at 24 months after start of treatment. The secondary endpoints include assessment of the following at 12, 24, and 36 months: composite risk score iBox, safety, evolution and characteristics of donor-specific antibodies (DSA), graft histology, proteinuria, kidney function assessed by measured GFR (mGFR), patient- and death-censored graft survival, and patient-reported outcomes that include transplant-specific well-being, adherence to immunosuppressive medications and perceived threat of the risk of graft rejection. DISCUSSION No effective treatment exists for caAMR at present. Based on the hypothesis that inhibition of IL-6 receptor by tocilizumab will reduce antibody production and reduce antibody-mediated damage, our randomized trial has a potential to provide evidence for a novel treatment strategy for caAMR, therewith slowing the decline in graft function in the long-term. TRIAL REGISTRATION ClinicalTrials.gov NCT04561986. Registered on September 24, 2020.
Collapse
Affiliation(s)
- Lillian Streichart
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Marie Felldin
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Jana Ekberg
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Lars Mjörnstedt
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Per Lindnér
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Annette Lennerling
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Verena Bröcker
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Mölne
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Holgersson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg and Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristien Daenen
- Department of Nephrology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Wennberg
- Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Lorant
- Section of Transplantation Surgery, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Seema Baid-Agrawal
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden.
| |
Collapse
|
11
|
Roufosse C, Naesens M, Haas M, Lefaucheur C, Mannon RB, Afrouzian M, Alachkar N, Aubert O, Bagnasco SM, Batal I, Bellamy COC, Broecker V, Budde K, Clahsen-Van Groningen M, Coley SM, Cornell LD, Dadhania D, Demetris AJ, Einecke G, Farris AB, Fogo AB, Friedewald J, Gibson IW, Horsfield C, Huang E, Husain SA, Jackson AM, Kers J, Kikić Ž, Klein A, Kozakowski N, Liapis H, Mangiola M, Montgomery RA, Nankinvell B, Neil DAH, Nickerson P, Rabant M, Randhawa P, Riella LV, Rosales I, Royal V, Sapir-Pichhadze R, Sarder P, Sarwal M, Schinstock C, Stegall M, Solez K, van der Laak J, Wiebe C, Colvin RB, Loupy A, Mengel M. The Banff 2022 Kidney Meeting Work Plan: Data-driven refinement of the Banff Classification for renal allografts. Am J Transplant 2024; 24:350-361. [PMID: 37931753 PMCID: PMC11135910 DOI: 10.1016/j.ajt.2023.10.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
The XVIth Banff Meeting for Allograft Pathology was held in Banff, Alberta, Canada, from September 19 to 23, 2022, as a joint meeting with the Canadian Society of Transplantation. In addition to a key focus on the impact of microvascular inflammation and biopsy-based transcript analysis on the Banff Classification, further sessions were devoted to other aspects of kidney transplant pathology, in particular T cell-mediated rejection, activity and chronicity indices, digital pathology, xenotransplantation, clinical trials, and surrogate endpoints. Although the output of these sessions has not led to any changes in the classification, the key role of Banff Working Groups in phrasing unanswered questions, and coordinating and disseminating results of investigations addressing these unanswered questions was emphasized. This paper summarizes the key Banff Meeting 2022 sessions not covered in the Banff Kidney Meeting 2022 Report paper and also provides an update on other Banff Working Group activities relevant to kidney allografts.
Collapse
Affiliation(s)
- Candice Roufosse
- Department of Immunology and Inflammation, Faculty Medicine, Imperial College London, London, UK.
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Carmen Lefaucheur
- Université Paris Cité, INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, France & Department of Nephrology and Transplantation, Saint-Louis Hospital, Paris, France
| | - Roslyn B Mannon
- Department of Internal Medicine, Division of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Marjan Afrouzian
- Department of Pathology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Nada Alachkar
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olivier Aubert
- Université Paris Cité, INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, France & Department of Transplantation, Necker Hospital, Paris, France
| | - Serena M Bagnasco
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ibrahim Batal
- Pathology & Cell Biology, Columbia University Irving Medical Center, New York, USA
| | | | - Verena Broecker
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Klemens Budde
- Department of Nephrology, Charité Universitätsmedizin, Berlin, Germany
| | - Marian Clahsen-Van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Center Rotterdam, Rotterdam, Netherlands; Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Shana M Coley
- Transplant Translational Research, Arkana Laboratories, Arkansas, USA
| | - Lynn D Cornell
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Darshana Dadhania
- Department Medicine, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Anthony J Demetris
- UPMC Hepatic and Transplantation Pathology, Pittsburg, Pennsylvania, USA
| | - Gunilla Einecke
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Germany
| | - Alton B Farris
- Department of Pathology and Laboratory Medicine, Emory University, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John Friedewald
- Comprehensive Transplant Center, Northwestern University, USA
| | - Ian W Gibson
- Department of Pathology, University of Manitoba, Winnipeg, Canada
| | | | - Edmund Huang
- Department of Medicine, Division of Nephrology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Syed A Husain
- Division of Nephrology, Columbia University, New York, New York, USA
| | | | - Jesper Kers
- Department of Pathology, Leiden University Medical Center, Netherlands; Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Željko Kikić
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | | | | | - Helen Liapis
- Ludwig Maximillian University Munich, Nephrology Center, Germany
| | | | | | - Brian Nankinvell
- Department of Renal Medicine, Westmead Hospital, Westmead, New South Wales, Australia
| | - Desley A H Neil
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham and Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Peter Nickerson
- Department of Medicine and Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Marion Rabant
- Pathology department, Necker-Enfants Malades Hospital, Paris, France
| | - Parmjeet Randhawa
- Pathology, Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Leonardo V Riella
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Rosales
- Immunopathology Research Laboratory, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Virginie Royal
- Maisonneuve-Rosemont Hospital, University of Montreal, Quebec, Canada
| | - Ruth Sapir-Pichhadze
- Division of Nephrology & Multiorgan Transplant Program, McGill University, Montreal, Quebec, Canada
| | - Pinaki Sarder
- Department of Medicine-Quantitative Health, University of Florida College of Medicine, Florida, USA
| | - Minnie Sarwal
- Division of MultiOrgan Transplantation, UCSF, San Francisco, California, USA
| | - Carrie Schinstock
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark Stegall
- Department Transplantation Surgery, Mayo Clinic, Rochester, Massachusetts, USA
| | - Kim Solez
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | | | - Chris Wiebe
- Department of Medicine and Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Robert B Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandre Loupy
- Université Paris Cité, INSERM, PARCC, Paris Institute for Transplantation and Organ Regeneration, France & Department of Transplantation, Necker Hospital, Paris, France
| | - Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| |
Collapse
|
12
|
Zhang D, Zhang H, Lu J, Hu X. Multiomics Data Reveal the Important Role of ANXA2R in T Cell-mediated Rejection After Renal Transplantation. Transplantation 2024; 108:430-444. [PMID: 37677931 PMCID: PMC10798590 DOI: 10.1097/tp.0000000000004754] [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] [Received: 04/17/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND T cell-mediated rejection (TCMR) is a severe issue after renal transplantation, but research on its T cell-receptor (TCR) repertoire is lacking. This study intended to elucidate the TCR repertoire landscape in TCMR and hence identify novel potential targets. METHODS A total of 12 multiomics data sets were collected. The TRUST4 algorithm was used to construct and analyze the TCR repertoire in renal allografts with TCMR and stable renal function. Then, novel TCR-related key genes were identified through various criteria and literature research. In bulk transcriptome, cell line, single-cell transcriptome data sets, multiple immune cell infiltration algorithms, and gene set enrichment analysis were used to analyze potential mechanisms of the identified key gene. Twenty-three pathological sections were collected for immunofluorescence staining in the clinical cohort. Finally, the diagnostic and prognostic values of ANXA2R were evaluated in multiple renal transplant data sets. RESULTS Allografts with TCMR showed significantly increased clonotype and specific clonal expansion. ANXA2R was found to be a novel key gene for TCMR and showed strong positive connections with the TCR complex and lymphocyte cells, especially CD8 + T cells. Immunofluorescence staining confirmed the existence of ANXA2R + CD8 + T cells, with their percentage significantly elevated in TCMR compared with stable renal function. Finally, both mRNA and protein levels of ANXA2R showed promising diagnostic and prognostic value for renal transplant recipients. CONCLUSIONS ANXA2R , identified as a novel TCR-related gene, had critical roles in clinicopathology, diagnosis, and prognosis in renal transplantation, which offered promising potential therapeutic targets.
Collapse
Affiliation(s)
- Di Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - He Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jun Lu
- Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
López Del Moral C, Wu K, Naik M, Osmanodja B, Akifova A, Lachmann N, Stauch D, Hergovits S, Choi M, Bachmann F, Halleck F, Schrezenmeier E, Schmidt D, Budde K. Predictors of graft failure after first detection of de novo donor-specific HLA antibodies in kidney transplant recipients. Nephrol Dial Transplant 2023; 39:84-94. [PMID: 37410616 DOI: 10.1093/ndt/gfad149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND De novo donor-specific antibodies (dnDSAs) may cause antibody-mediated rejection and graft dysfunction. Little is known about the clinical course after first detection of dnDSAs during screening in asymptomatic patients. We aimed to assess the value of estimated glomerular filtration rate (eGFR) and proteinuria to predict graft failure in patients with dnDSAs and their potential utility as surrogate endpoints. METHODS All 400 kidney transplant recipients with dnDSAs at our centre (1 March 2000-31 May 2021) were included in this retrospective study. The dates of graft loss, rejection, doubling of creatinine, ≥30% eGFR decline, proteinuria ≥500 mg/g and ≥1000 mg/g were registered from the first dnDSA appearance. RESULTS During 8.3 years of follow-up, graft failure occurred in 33.3% of patients. Baseline eGFR and proteinuria correlated with 5-year graft loss (area under the receiver operating characteristics curve 0.75 and 0.80, P < .001). Creatinine doubled after a median of 2.8 years [interquartile range (IQR) 1.5-5.0] from dnDSA and the time from doubling creatinine to graft failure was 1.0 year (IQR 0.4-2.9). Analysing eGFR reduction ≥30% as a surrogate endpoint (148/400), the time from dnDSA to this event was 2.0 years (IQR 0.6-4.2), with a positive predictive value (PPV) of 45.9% to predict graft loss, which occurred after 2.0 years (IQR 0.8-3.2). The median time from proteinuria ≥500 mg/g and ≥1000 mg/g to graft failure was identical, 1.8 years, with a PPV of 43.8% and 49.0%, respectively. Composite endpoints did not improve PPV. Multivariable analysis showed that rejection was the most important independent risk factor for all renal endpoints and graft loss. CONCLUSIONS Renal function, proteinuria and rejection are strongly associated with graft failure in patients with dnDSA and may serve as surrogate endpoints.
Collapse
Affiliation(s)
- Covadonga López Del Moral
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Nephrology, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain
| | - Kaiyin Wu
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel Naik
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aylin Akifova
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Lachmann
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Diana Stauch
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sabine Hergovits
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health Charité - Universitätsmedizin Berlin, BIH Academy, Berlin, Germany
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
14
|
Josephson MA, Becker Y, Budde K, Kasiske BL, Kiberd BA, Loupy A, Małyszko J, Mannon RB, Tönshoff B, Cheung M, Jadoul M, Winkelmayer WC, Zeier M. Challenges in the management of the kidney allograft: from decline to failure: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int 2023; 104:1076-1091. [PMID: 37236423 DOI: 10.1016/j.kint.2023.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
In March 2022, Kidney Disease: Improving Global Outcomes (KDIGO) held a virtual Controversies Conference to address the important but rarely examined phase during which the kidney transplant is failing or has failed. In addition to discussing the definition of a failing allograft, 4 broad areas were considered in the context of a declining functioning graft: prognosis and kidney failure trajectory; immunosuppression strategies; management of medical and psychological complications, and patient factors; and choice of kidney replacement therapy or supportive care following graft loss. Identifying and paying special attention to individuals with failing allografts was felt to be important in order to prepare patients psychologically, manage immunosuppression, address complications, prepare for dialysis and/or retransplantation, and transition to supportive care. Accurate prognostication tools, although not yet widely available, were embraced as necessary to define allograft survival trajectories and the likelihood of allograft failure. The decision of whether to withdraw or continue immunosuppression after allograft failure was deemed to be based most appropriately on risk-benefit analysis and likelihood of retransplantation within a few months. Psychological preparation and support was identified as a critical factor in patient adjustment to graft failure, as was early communication. Several models of care were noted that enabled a medically supportive transition back to dialysis or retransplantation. Emphasis was placed on the importance of dialysis-access readiness before initiation of dialysis, in order to avoid use of central venous catheters. The centrality of the patient to all management decisions and discussions was deemed to be paramount. Patient "activation," which can be defined as engaged agency, was seen as the most effective way to achieve success. Unresolved controversies, gaps in knowledge, and areas for research were also stressed in the conference deliberations.
Collapse
Affiliation(s)
- Michelle A Josephson
- Section of Nephrology, Department of Medicine, and Transplant Institute, University of Chicago, Chicago, Illinois, USA.
| | - Yolanda Becker
- Transplantation Institute, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Bertram L Kasiske
- Department of Medicine, Hennepin Healthcare, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bryce A Kiberd
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alexandre Loupy
- Université Paris Cité, INSERM U970, Paris Institute for Transplantation and Organ Regeneration, F-75015 Paris, France; Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Roslyn B Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Michael Cheung
- Kidney Disease: Improving Global Outcomes (KDIGO), Brussels, Belgium
| | - Michel Jadoul
- Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Wolfgang C Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Martin Zeier
- Division of Nephrology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
15
|
Saben JL, Schold JD, Kaplan B. The Use of In Silico and Mathematical Modeling to Create More Accurate and Efficient Clinical Trial Design. Transplantation 2023; 107:2292-2293. [PMID: 37870881 DOI: 10.1097/tp.0000000000004733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Affiliation(s)
- Jessica L Saben
- Department of Surgery, Colorado Center for Transplantation Care, Research, and Education (CCTCARE), University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jesse D Schold
- Department of Surgery, Colorado Center for Transplantation Care, Research, and Education (CCTCARE), University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Bruce Kaplan
- Department of Medicine, Colorado Center for Transplantation Care, Research, and Education (CCTCARE), University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
16
|
Naesens M. Embracing the Wisdom of Ancient Greece in the Era of Personalized Medicine-Uncertainty, Probabilistic Reasoning, and Democratic Consensus. Transpl Int 2023; 36:12178. [PMID: 37954528 PMCID: PMC10632184 DOI: 10.3389/ti.2023.12178] [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: 10/03/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023]
Abstract
Further improvements of outcome after solid organ transplantation will depend on our ability to integrate personalized medicine in clinical routine. Not only better risk stratification or improved diagnostics, also targeted therapies and predictive markers of treatment success are needed, as there is a virtual standstill in the development and implementation of novel therapies for prevention and treatment of allograft rejection. The integration of clinical decision support algorithms and novel biomarkers in clinical practice will require a different reasoning, embracing concepts of uncertainty and probabilistic thinking as the ground truth is often unknown and the tools imperfect. This is important for communication between healthcare professionals, but patients and their caregivers also need to be informed and educated about the levels of uncertainty inherent to personalized medicine. In the translation of research findings and personalized medicine to routine clinical care, it remains crucial to maintain global consensus on major aspects of clinical routine, to avoid further divergence between centres and countries in the standard of care. Such consensus can only be reached when experts with divergent opinions are willing to transcend their own convictions, understand that there is not one single truth, and thus are able to embrace a level of uncertainty.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Heldal TF, Åsberg A, Ueland T, Reisæter AV, Pischke SE, Mollnes TE, Aukrust P, Reinholt F, Hartmann A, Heldal K, Jenssen TG. Systemic inflammation early after kidney transplantation is associated with long-term graft loss: a cohort study. Front Immunol 2023; 14:1253991. [PMID: 37849758 PMCID: PMC10577420 DOI: 10.3389/fimmu.2023.1253991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
Background Early graft loss following kidney transplantation is mainly a result of acute rejection or surgical complications, while long-term kidney allograft loss is more complex. We examined the association between systemic inflammation early after kidney transplantation and long-term graft loss, as well as correlations between systemic inflammation scores and inflammatory findings in biopsies 6 weeks and 1 year after kidney transplantation. Methods We measured 21 inflammatory biomarkers 10 weeks after transplantation in 699 patients who were transplanted between 2009 and 2012 at Oslo University Hospital, Rikshospitalet, Norway. Low-grade inflammation was assessed with predefined inflammation scores based on specific biomarkers: one overall inflammation score and five pathway-specific scores. Surveillance or indication biopsies were performed in all patients 6 weeks after transplantation. The scores were tested in Cox regression models. Results Median follow-up time was 9.1 years (interquartile range 7.6-10.7 years). During the study period, there were 84 (12.2%) death-censored graft losses. The overall inflammation score was associated with long-term kidney graft loss both when assessed as a continuous variable (hazard ratio 1.03, 95% CI 1.01-1.06, P = 0.005) and as a categorical variable (4th quartile: hazard ratio 3.19, 95% CI 1.43-7.10, P = 0.005). In the pathway-specific analyses, fibrogenesis activity and vascular inflammation stood out. The vascular inflammation score was associated with inflammation in biopsies 6 weeks and 1 year after transplantation, while the fibrinogenesis score was associated with interstitial fibrosis and tubular atrophy. Conclusion In conclusion, a systemic inflammatory environment early after kidney transplantation was associated with biopsy-confirmed kidney graft pathology and long-term kidney graft loss. The systemic vascular inflammation score correlated with inflammatory findings in biopsies 6 weeks and 1 year after transplantation.
Collapse
Affiliation(s)
- Torbjørn F. Heldal
- Department of Internal Medicine, Telemark Hospital Trust, Skien, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Anders Åsberg
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Norwegian Renal Registry, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
- Research Institute of Internal Medicine, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Anna V. Reisæter
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Norwegian Renal Registry, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Søren E. Pischke
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Anesthesiology, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Tom E. Mollnes
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway
| | - Pål Aukrust
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
- Research Institute of Internal Medicine, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Finn Reinholt
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anders Hartmann
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| | - Kristian Heldal
- Department of Internal Medicine, Telemark Hospital Trust, Skien, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
- Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Trond G. Jenssen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital – Rikshospitalet, Oslo, Norway
| |
Collapse
|
18
|
Hogan J, Divard G, Aubert O, Garro R, Boyer O, Donald Cooper LA, Farris AB, Fila M, Seifert M, Sellier-Leclerc AL, Smith J, Fichtner A, Tönshoff B, Twombley K, Warady B, Pearl M, Zahr RS, Lefaucheur C, Patzer R, Loupy A. Validation of a prediction system for risk of kidney allograft failure in pediatric kidney transplant recipients: An international observational study. Am J Transplant 2023; 23:1561-1569. [PMID: 37453485 PMCID: PMC11247401 DOI: 10.1016/j.ajt.2023.07.004] [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: 01/13/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Predicting long-term kidney allograft failure is an unmet need for clinical care and clinical trial optimization in children. We aimed to validate a kidney allograft failure risk prediction system in a large international cohort of pediatric kidney transplant recipients. Patients from 20 centers in Europe and the United States, transplanted between 2004 and 2017, were included. Allograft assessment included estimated glomerular filtration rate, urine protein-to-creatinine ratio, circulating antihuman leukocyte antigen donor-specific antibody, and kidney allograft histology. Individual predictions of allograft failure were calculated using the integrative box (iBox) system. Prediction performances were assessed using discrimination and calibration. The allograft evaluations were performed in 706 kidney transplant recipients at a median time of 9.1 (interquartile range, 3.3-19.2) months posttransplant; mean estimated glomerular filtration rate was 68.7 ± 28.1 mL/min/1.73 m2, and median urine protein-to-creatinine ratio was 0.1 (0.0-0.4) g/g, and 134 (19.0%) patients had antihuman leukocyte antigen donor-specific antibodies. The iBox exhibited accurate calibration and discrimination for predicting the outcomes up to 10 years after evaluation, with a C-index of 0.81 (95% confidence interval, 0.75-0.87). This study confirms the generalizability of the iBox to predict long-term kidney allograft failure in children, with performances similar to those reported in adults. These results support the use of the iBox to improve patient monitoring and facilitate clinical trials in children.
Collapse
Affiliation(s)
- Julien Hogan
- Université Paris Cité, INSERM, UMR-S970, PARCC, Paris Translational Research Center for Organ Transplantation, Paris, France; Pediatric nephrology department, Robert Debré Hospital, APHP, Paris, France; Emory Transplant Center, Department of Surgery, Emory University, Atlanta, Georgia, USA
| | - Gillian Divard
- Université Paris Cité, INSERM, UMR-S970, PARCC, Paris Translational Research Center for Organ Transplantation, Paris, France
| | - Olivier Aubert
- Université Paris Cité, INSERM, UMR-S970, PARCC, Paris Translational Research Center for Organ Transplantation, Paris, France
| | - Rouba Garro
- Pediatric Nephrology Department, Children Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Olivia Boyer
- Pediatric Nephrology, MARHEA Reference Center, INSERM U1163, Imagine Institute, Paris Cité University, Necker-Enfants Malades Hospital, APHP.Centre, Paris, France
| | - Lee Alex Donald Cooper
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alton Brad Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marc Fila
- Pediatric Nephrology Department, Montpellier University Hospital, Montpellier, France
| | - Michael Seifert
- Pediatric Nephrology Department, University of Alabama, Birmingham, Alabama, USA
| | | | - Jody Smith
- Pediatric Nephrology Department, Seattle Children, Seattle, New York, USA
| | - Alexander Fichtner
- Department of Pediatrics I, University Childrens Hospital Heidelberg, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Childrens Hospital Heidelberg, Heidelberg, Germany
| | - Katherine Twombley
- Pediatric Nephrology Department, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bradley Warady
- Pediatric Nephrology Department, Children's Mercy, Kansas City, Michigan, USA
| | - Meghan Pearl
- Pediatric Nephrology Department, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Rima S Zahr
- UTHSC Department of Pediatric Nephrology and Hypertension, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Carmen Lefaucheur
- Université Paris Cité, INSERM, UMR-S970, PARCC, Paris Translational Research Center for Organ Transplantation, Paris, France
| | - Rachel Patzer
- Emory Transplant Center, Department of Surgery, Emory University, Atlanta, Georgia, USA
| | - Alexandre Loupy
- Université Paris Cité, INSERM, UMR-S970, PARCC, Paris Translational Research Center for Organ Transplantation, Paris, France.
| |
Collapse
|
19
|
Sypek M, Francis A, Chadban S. Predicting graft survival in pediatric kidney transplantation: Does the Box fit? Am J Transplant 2023; 23:1481-1482. [PMID: 37652175 DOI: 10.1016/j.ajt.2023.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Affiliation(s)
- Matthew Sypek
- Department of Nephrology, Royal Melbourne Hospital, Victoria, Australia; Department of Nephrology, Royal Children's Hospital, Victoria, Australia; Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Victoria, Australia
| | - Anna Francis
- Department of Nephrology, Queensland Children's Hospital, Queensland, Australia
| | - Steve Chadban
- Renal Medicine, Royal Prince Alfred Hospital, New South Wales, Australia; Kidney Node, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Australia.
| |
Collapse
|
20
|
Budde K, Kaplan B. Stronger together: Lessons from the iBox qualification process. Am J Transplant 2023; 23:1478-1480. [PMID: 37236402 DOI: 10.1016/j.ajt.2023.05.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Affiliation(s)
- Klemens Budde
- Department of Nephrology,Charité Universitätsmedizin Berlin,Berlin,Germany
| | - Bruce Kaplan
- Department of Medicine,Colorado Center for Transplantation Care,Research and Education (CCTCARE),University of Colorado,Anschutz Medical Campus,Aurora,Colorado,USA.
| |
Collapse
|
21
|
Klein A, Loupy A, Stegall M, Helanterä I, Kosinski L, Frey E, Aubert O, Divard G, Newell K, Meier-Kriesche HU, Mannon RB, Dumortier T, Aggarwal V, Podichetty JT, O'Doherty I, Gaber AO, Fitzsimmons WE. Qualifying a novel clinical trial endpoint (iBOX) predictive of long-term kidney transplant outcomes. Am J Transplant 2023; 23:1496-1506. [PMID: 37735044 DOI: 10.1016/j.ajt.2023.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 09/23/2023]
Abstract
New immunosuppressive therapies that improve long-term graft survival are needed in kidney transplant. Critical Path Institute's Transplant Therapeutics Consortium received a qualification opinion for the iBOX Scoring System as a novel secondary efficacy endpoint for kidney transplant clinical trials through European Medicines Agency's qualification of novel methodologies for drug development. This is the first qualified endpoint for any transplant indication and is now available for use in kidney transplant clinical trials. Although the current efficacy failure endpoint has typically shown the noninferiority of therapeutic regimens, the iBOX Scoring System can be used to demonstrate the superiority of a new immunosuppressive therapy compared to the standard of care from 6 months to 24 months posttransplant in pivotal or exploratory drug therapeutic studies.
Collapse
Affiliation(s)
| | - Alexandre Loupy
- Université de Paris, Cité, Institut national de la santé et de la recherche médicale, U970, PARCC, Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Mark Stegall
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ilkka Helanterä
- Department of Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | | | - Eric Frey
- Critical Path Institute, Tucson, Arizona, USA
| | - Olivier Aubert
- Université de Paris, Cité, Institut national de la santé et de la recherche médicale, U970, PARCC, Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Gillian Divard
- Université de Paris, Cité, Institut national de la santé et de la recherche médicale, U970, PARCC, Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Kenneth Newell
- Division of Transplantation, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Roslyn B Mannon
- Department of Medicine, Division of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | - Ahmed Osama Gaber
- Department of Surgery, Houston Methodist Hospital, Houston, Texas, USA, and Weill Cornell Medicine, New York, New York, USA
| | | |
Collapse
|
22
|
Klein A, Loupy A, Stegall M, Helanterä I, Kosinski L, Frey E, Aubert O, Divard G, Newell K, Meier-Kriesche HU, Mannon R, Dumortier T, Aggarwal V, Podichetty JT, O’Doherty I, Gaber AO, Fitzsimmons WE. Qualifying a Novel Clinical Trial Endpoint (iBOX) Predictive of Long-Term Kidney Transplant Outcomes. Transpl Int 2023; 36:11951. [PMID: 37822449 PMCID: PMC10563802 DOI: 10.3389/ti.2023.11951] [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: 12/30/2022] [Accepted: 04/12/2023] [Indexed: 10/13/2023]
Abstract
New immunosuppressive therapies that improve long-term graft survival are needed in kidney transplant. Critical Path Institute's Transplant Therapeutics Consortium received a qualification opinion for the iBOX Scoring System as a novel secondary efficacy endpoint for kidney transplant clinical trials through European Medicines Agency's qualification of novel methodologies for drug development. This is the first qualified endpoint for any transplant indication and is now available for use in kidney transplant clinical trials. Although the current efficacy failure endpoint has typically shown the noninferiority of therapeutic regimens, the iBOX Scoring System can be used to demonstrate the superiority of a new immunosuppressive therapy compared to the standard of care from 6 months to 24 months posttransplant in pivotal or exploratory drug therapeutic studies.
Collapse
Affiliation(s)
- Amanda Klein
- Critical Path Institute, Tucson, AZ, United States
| | - Alexandre Loupy
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Mark Stegall
- Department of Surgery, Mayo Clinic, Rochester, Rochester, MN, United States
| | - Ilkka Helanterä
- Department of Transplantation and Liver Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Eric Frey
- Critical Path Institute, Tucson, AZ, United States
| | - Olivier Aubert
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Gillian Divard
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Kenneth Newell
- Division of Transplantation, Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | | | - Roslyn Mannon
- Division of Nephrology, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | | | | | | | | | - Ahmed Osama Gaber
- Department of Surgery, Houston Methodist Hospital, Houston, TX, United States
| | | |
Collapse
|
23
|
Vaulet T, Divard G, Thaunat O, Koshy P, Lerut E, Senev A, Aubert O, Van Loon E, Callemeyn J, Emonds MP, Van Craenenbroeck A, De Vusser K, Sprangers B, Rabeyrin M, Dubois V, Kuypers D, De Vos M, Loupy A, De Moor B, Naesens M. Data-Driven Chronic Allograft Phenotypes: A Novel and Validated Complement for Histologic Assessment of Kidney Transplant Biopsies. J Am Soc Nephrol 2022; 33:2026-2039. [PMID: 36316096 PMCID: PMC9678036 DOI: 10.1681/asn.2022030290] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/24/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND No validated system currently exists to realistically characterize the chronic pathology of kidney transplants that represents the dynamic disease process and spectrum of disease severity. We sought to develop and validate a tool to describe chronicity and severity of renal allograft disease and integrate it with the evaluation of disease activity. METHODS The training cohort included 3549 kidney transplant biopsies from an observational cohort of 937 recipients. We reweighted the chronic histologic lesions according to their time-dependent association with graft failure, and performed consensus k-means clustering analysis. Total chronicity was calculated as the sum of the weighted chronic lesion scores, scaled to the unit interval. RESULTS We identified four chronic clusters associated with graft outcome, based on the proportion of ambiguous clustering. The two clusters with the worst survival outcome were determined by interstitial fibrosis and tubular atrophy (IFTA) and by transplant glomerulopathy. The chronic clusters partially overlapped with the existing Banff IFTA classification (adjusted Rand index, 0.35) and were distributed independently of the acute lesions. Total chronicity strongly associated with graft failure (hazard ratio [HR], 8.33; 95% confidence interval [CI], 5.94 to 10.88; P<0.001), independent of the total activity scores (HR, 5.01; 95% CI, 2.83 to 7.00; P<0.001). These results were validated on an external cohort of 4031 biopsies from 2054 kidney transplant recipients. CONCLUSIONS The evaluation of total chronicity provides information on kidney transplant pathology that complements the estimation of disease activity from acute lesion scores. Use of the data-driven algorithm used in this study, called RejectClass, may provide a holistic and quantitative assessment of kidney transplant injury phenotypes and severity.
Collapse
Affiliation(s)
- Thibaut Vaulet
- ESAT Stadius Center for Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Leuven, Belgium
| | - Gillian Divard
- Paris Translational Research Center for Organ Transplantation, Université de Paris, INSERM, PARCC, Paris, France; Kidney Transplant Department, Necker Hospital, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology, and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Priyanka Koshy
- Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Evelyne Lerut
- Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Aleksandar Senev
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross–Flanders, Mechelen, Belgium
| | - Olivier Aubert
- Paris Translational Research Center for Organ Transplantation, Université de Paris, INSERM, PARCC, Paris, France; Kidney Transplant Department, Necker Hospital, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Elisabet Van Loon
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Jasper Callemeyn
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Marie-Paule Emonds
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross–Flanders, Mechelen, Belgium
| | - Amaryllis Van Craenenbroeck
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Katrien De Vusser
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | - Valérie Dubois
- Human Leukocyte Antigen (HLA) Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Dirk Kuypers
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Maarten De Vos
- ESAT Stadius Center for Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Université de Paris, INSERM, PARCC, Paris, France; Kidney Transplant Department, Necker Hospital, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Bart De Moor
- ESAT Stadius Center for Dynamical Systems, Signal Processing, and Data Analytics, KU Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Affiliation(s)
- Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Leuven Biostatistics and Statistical Bioinformatics Centre (L-Biostat), KU Leuven, Leuven, Belgium
| | - Geert Verbeke
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-Biostat), Universiteit Hasselt and KU Leuven, Hasselt and Leuven, Belgium
| | - Bernd Döhler
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Caner Süsal
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
- Transplant Immunology Research Centre of Excellence, Koç University, Istanbul, Turkey
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Hilbrands L, Budde K, Bellini MI, Diekmann F, Furian L, Grinyó J, Heemann U, Hesselink DA, Loupy A, Oberbauer R, Pengel L, Reinders M, Schneeberger S, Naesens M. Allograft Function as Endpoint for Clinical Trials in Kidney Transplantation. Transpl Int 2022; 35:10139. [PMID: 35669976 PMCID: PMC9163811 DOI: 10.3389/ti.2022.10139] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/11/2022] [Indexed: 12/14/2022]
Abstract
Clinical study endpoints that assess the efficacy of interventions in patients with chronic renal insufficiency can be adopted for use in kidney transplantation trials, given the pathophysiological similarities between both conditions. Kidney dysfunction is reflected in the glomerular filtration rate (GFR), and although a predefined (e.g., 50%) reduction in GFR was recommended as an endpoint by the European Medicines Agency (EMA) in 2016, many other endpoints are also included in clinical trials. End-stage renal disease is strongly associated with a change in estimated (e)GFR, and eGFR trajectories or slopes are increasingly used as endpoints in clinical intervention trials in chronic kidney disease (CKD). Similar approaches could be considered for clinical trials in kidney transplantation, although several factors should be taken into account. The present Consensus Report was developed from documentation produced by the European Society for Organ Transplantation (ESOT) as part of a Broad Scientific Advice request that ESOT submitted to the EMA in 2020. This paper provides a contemporary discussion of primary endpoints used in clinical trials involving CKD, including proteinuria and albuminuria, and evaluates the validity of these concepts as endpoints for clinical trials in kidney transplantation.
Collapse
Affiliation(s)
- Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Fritz Diekmann
- Department of Nephrology and Kidney Transplantation, Vall d’Hebrón University Hospital, Barcelona, Spain
| | - Lucrezia Furian
- Kidney and Pancreas Transplantation Unit, University of Padua, Padua, Italy
| | - Josep Grinyó
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Dennis A. Hesselink
- Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | - Liset Pengel
- Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Marlies Reinders
- Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan Schneeberger
- Department of General, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| |
Collapse
|
26
|
Naesens M, Loupy A, Hilbrands L, Oberbauer R, Bellini MI, Glotz D, Grinyó J, Heemann U, Jochmans I, Pengel L, Reinders M, Schneeberger S, Budde K. Rationale for Surrogate Endpoints and Conditional Marketing Authorization of New Therapies for Kidney Transplantation. Transpl Int 2022; 35:10137. [PMID: 35669977 PMCID: PMC9163307 DOI: 10.3389/ti.2022.10137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022]
Abstract
Conditional marketing authorization (CMA) facilitates timely access to new drugs for illnesses with unmet clinical needs, such as late graft failure after kidney transplantation. Late graft failure remains a serious, burdensome, and life-threatening condition for recipients. This article has been developed from content prepared by members of a working group within the European Society for Organ Transplantation (ESOT) for a Broad Scientific Advice request, submitted by ESOT to the European Medicines Agency (EMA), and reviewed by the EMA in 2020. The article presents the rationale for using surrogate endpoints in clinical trials aiming at improving late graft failure rates, to enable novel kidney transplantation therapies to be considered for CMA and improve access to medicines. The paper also provides background data to illustrate the relationship between primary and surrogate endpoints. Developing surrogate endpoints and a CMA strategy could be particularly beneficial for studies where the use of primary endpoints would yield insufficient statistical power or insufficient indication of long-term benefit following transplantation.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| | - Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | | | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, Hôpital Saint Louis, Paris, France
| | | | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Ina Jochmans
- Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Liset Pengel
- Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Marlies Reinders
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan Schneeberger
- Department of General, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
27
|
Deville KA, Seifert ME. Biomarkers of alloimmune events in pediatric kidney transplantation. Front Pediatr 2022; 10:1087841. [PMID: 36741087 PMCID: PMC9895094 DOI: 10.3389/fped.2022.1087841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023] Open
Abstract
Alloimmune events such as the development of de novo donor-specific antibody (dnDSA), T cell-mediated rejection (TCMR), and antibody-mediated rejection (ABMR) are the primary contributors to kidney transplant failure in children. For decades, a creatinine-based estimated glomerular filtration rate (eGFR) has been the non-invasive gold standard biomarker for detecting clinically significant alloimmune events, but it suffers from low sensitivity and specificity, especially in smaller children and older allografts. Many clinically "stable" children (based on creatinine) will have alloimmune events known as "subclinical acute rejection" (based on biopsy) that merely reflect the inadequacy of creatinine-based estimates for alloimmune injury rather than a distinct phenotype from clinical rejection with allograft dysfunction. The poor biomarker performance of creatinine leads to many unnecessary surveillance and for-cause biopsies that could be avoided by integrating non-invasive biomarkers with superior sensitivity and specificity into current clinical paradigms. In this review article, we will present and appraise the current state-of-the-art in monitoring for alloimmune events in pediatric kidney transplantation. We will first discuss the current clinical standards for assessing the presence of alloimmune injury and predicting long-term outcomes. We will review principles of biomarker medicine and the application of comprehensive metrics to assess the performance of a given biomarker against the current gold standard. We will then highlight novel blood- and urine-based biomarkers (with special emphasis on pediatric biomarker studies) that have shown superior diagnostic and prognostic performance to the current clinical standards including creatinine-based eGFR. Finally, we will review some of the barriers to translating this research and implementing emerging biomarkers into common clinical practice, and present a transformative approach to using multiple biomarker platforms at different times to optimize the detection and management of critical alloimmune events in pediatric kidney transplant recipients.
Collapse
Affiliation(s)
- Kyle A Deville
- Division of Pediatric Nephrology, Department of Pediatrics, University of Alabama Heersink School of Medicine, Birmingham, AL, United States
| | - Michael E Seifert
- Division of Pediatric Nephrology, Department of Pediatrics, University of Alabama Heersink School of Medicine, Birmingham, AL, United States
| |
Collapse
|