1
|
Mulley WR, Hughes PD, Collins MG, Pilmore HL, Clayton PA, Wyld ML, Lee D, van der Jeugd J, Fernando SC, Kuo SF, Tan S, Jahan S, Lim WH. Defining causes of death-censored kidney allograft failure: A 5-year multicentre ANZDATA and clinical cross-sectional study. Nephrology (Carlton) 2024; 29:930-940. [PMID: 39349052 PMCID: PMC11579561 DOI: 10.1111/nep.14397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/08/2024] [Accepted: 09/14/2024] [Indexed: 10/02/2024]
Abstract
AIM Determining specific causes of allograft failure allows a focus on understanding and treating these conditions. Previous studies highlight chronic antibody-mediated rejection as a leading cause of late allograft failure. We sought to define causes of allograft failure in a large cohort of kidney transplant recipients across multiple centres in Australia and New Zealand, including cases previously attributed to chronic allograft nephropathy (CAN). METHODS All death-censored allograft failures at 9 participating centres between 1 January 2014 to 31 December 2018 were included. Available clinical and biopsy data were reviewed and the "most likely" cause assigned. RESULTS There were 642 death-censored allograft failures in the study period. Of these, 495 (77.1%) had an informative biopsy performed a median of 13.4 months (IQR 2.5-39.1 months) prior to allograft failure. Rejection of any type was the leading cause of allograft failure (47.5%), comprised chiefly of chronic antibody-mediated rejection (37.4%) and chronic T-cell mediated rejection (6.4%). Other leading causes were undifferentiated interstitial fibrosis and tubular atrophy (10.8%), late medical and surgical complications (8.1%) and recurrent or de novo glomerulonephritis (7.0%). Polyoma viral nephropathy and calcineurin inhibitor toxicity each contributed to <2%. Causes of allograft failure previously attributed to CAN (n = 419, 65.3%) had a similar distribution to the overall cohort, with 43.9% attributed to chronic antibody-mediated rejection. CONCLUSION To prolong allograft survival, improved strategies are needed to curtail alloimmune responses. Greater understanding of the causes of undifferentiated interstitial fibrosis and tubular atrophy and potential treatments would also be of considerable benefit.
Collapse
Affiliation(s)
- William R. Mulley
- Department of NephrologyMonash Medical CentreClaytonVictoriaAustralia
- Centre for Inflammatory Diseases, Department of MedicineMonash UniversityClaytonVictoriaAustralia
| | - Peter D. Hughes
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of MedicineThe University of MelbourneMelbourneVictoriaAustralia
| | - Michael G. Collins
- Department of Renal MedicineAuckland City HospitalAucklandNew Zealand
- Central Northern Adelaide Renal and Transplantation ServiceRoyal Adelaide HospitalAdelaideAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideAustralia
| | - Helen L. Pilmore
- Department of Renal MedicineAuckland City HospitalAucklandNew Zealand
- Department of MedicineUniversity of AucklandAucklandNew Zealand
| | - Philip A. Clayton
- Central Northern Adelaide Renal and Transplantation ServiceRoyal Adelaide HospitalAdelaideAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideAustralia
- Australia & New Zealand Dialysis and Transplant (ANZDATA) RegistryAdelaideAustralia
| | - Melanie L. Wyld
- Department of Renal and Transplant MedicineWestmead HospitalWestmeadNew South WalesAustralia
- School of Public Health, Faculty of Medicine and HealthUniversity of SydneyCamperdownNew South WalesAustralia
| | - Darren Lee
- Department of Renal MedicineEastern HealthBox HillVictoriaAustralia
- Eastern Health Clinical SchoolMonash UniversityClaytonVictoriaAustralia
- Department of NephrologyAustin HealthHeidelbergVictoriaAustralia
| | | | - Sanduni C. Fernando
- Department of NephrologyMonash Medical CentreClaytonVictoriaAustralia
- Centre for Inflammatory Diseases, Department of MedicineMonash UniversityClaytonVictoriaAustralia
| | - Stephanie Fang‐Tzu Kuo
- Department of NephrologyThe Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of MedicineThe University of MelbourneMelbourneVictoriaAustralia
| | - Sarah Tan
- Department of NephrologyFlinders Medical CentreAdelaideAustralia
| | - Sadia Jahan
- Central Northern Adelaide Renal and Transplantation ServiceRoyal Adelaide HospitalAdelaideAustralia
| | - Wai H. Lim
- Department of Renal MedicineSir Charles Gairdner HospitalPerthAustralia
- Medical SchoolUniversity of Western AustraliaPerthAustralia
| |
Collapse
|
2
|
Cremoni M, Teisseyre M, Thaunat O, Fernandez C, Payre C, Moutou A, Zarif H, Brglez V, Albano L, Moal V, Mourad G, Morelon E, Hurault de Ligny B, Zaoui P, Rondeau E, Ouali N, Ronco P, Moulin B, Braun-Parvez L, Durrbach A, Heng AE, Grimbert P, Ducloux D, Blancho G, Merville P, Choukroun G, Le Meur Y, Vigneau C, Mariat C, Rostaing L, Subra JF, Taupin JL, Lambeau G, Esnault V, Sicard A, Seitz-Polski B. Anti Phospholipase A2 Receptor 1 Antibodies and Membranous Nephropathy Recurrence After Kidney Transplantation. Kidney Int Rep 2024; 9:3427-3438. [PMID: 39698349 PMCID: PMC11652070 DOI: 10.1016/j.ekir.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction Membranous nephropathy can lead to end-stage kidney disease, for which kidney transplantation is the preferred therapy. However, the disease often relapses, which can impact allograft survival. Methods We conducted a prospective multicenter study in France involving 72 patients with membranous nephropathy who were awaiting and then underwent kidney transplantation. In addition, we established a retrospective validation cohort of 65 patients. The primary objective was to evaluate the prognostic significance of pretransplant anti phospholipase A2 receptor 1 (PLA2R1) antibodies on the recurrence of membranous nephropathy. The study also assessed the incidence rate, time to onset, and risk factors for recurrence, as well as allograft outcome. Results The prospective cohort showed a 26% cumulative incidence of membranous nephropathy recurrence after a median follow-up of 23.5 months. This was confirmed by a 28% cumulative incidence after a median follow-up of 67 months in the retrospective cohort. A strong association was found between the presence of anti-PLA2R1 antibodies prior to transplantation and the risk of disease recurrence (risk ratio = 5.9; 95% confidence interval [CI]: 2.3-15.7; P < 0.0001). These results were confirmed in the retrospective cohort. Monitoring of anti-PLA2R1 antibodies in the immediate posttransplant period is of limited value, because recurrence occurred early in the first 6 months (median delay of 5 [3-14] months) after transplantation despite decreasing antibody levels. Conclusion The presence of anti-PLA2R1 antibodies prior to transplantation was a strong predictor of recurrence of allograft membranous nephropathy. An individualized immunomonitoring and management strategy for kidney transplant candidates with anti-PLA2R1-associated membranous nephropathy should be considered.
Collapse
Affiliation(s)
- Marion Cremoni
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, Centre Hospitier Universitaire de Nice, Nice, France
- Unité de Recherche Clinique Côte d'Azur (UR2CA), Université Côte d'Azur, Nice, France
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Pasteur 2, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Maxime Teisseyre
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, Centre Hospitier Universitaire de Nice, Nice, France
- Unité de Recherche Clinique Côte d'Azur (UR2CA), Université Côte d'Azur, Nice, France
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Pasteur 2, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Céline Fernandez
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, Centre Hospitier Universitaire de Nice, Nice, France
- Unité de Recherche Clinique Côte d'Azur (UR2CA), Université Côte d'Azur, Nice, France
| | - Christine Payre
- Institute of Molecular and Cellular Pharmacology, National Center for Scientific Research, University Côte d’Azur, UMR7275, Valbonne Sophia Antipolis, France
| | - Alan Moutou
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Hadi Zarif
- Institute of Molecular and Cellular Pharmacology, National Center for Scientific Research, University Côte d’Azur, UMR7275, Valbonne Sophia Antipolis, France
| | - Vesna Brglez
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, Centre Hospitier Universitaire de Nice, Nice, France
| | - Laetitia Albano
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Pasteur 2, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Valérie Moal
- Centre de Néphrologie et Transplantation Rénale, Aix Marseille Université, APHM, Hôpital Conception, Marseille, France
| | - Georges Mourad
- Department of Nephrology, Dialysis and Transplantation, Montpellier University hospital, Montpellier, France
| | - Emmanuel Morelon
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | | | - Philippe Zaoui
- Service de Néphrologie, Hémodialyse, Aphérèses et Transplantation Rénale, Grenoble Alpes University Hospital, La Tronche, France
| | - Eric Rondeau
- Nephrology Intensive Care, Department of Nephrology, Tenon Hospital, AP-HP, Paris, France
| | - Nacera Ouali
- Nephrology Intensive Care, Department of Nephrology, Tenon Hospital, AP-HP, Paris, France
| | - Pierre Ronco
- Nephrology Intensive Care, Department of Nephrology, Tenon Hospital, AP-HP, Paris, France
| | - Bruno Moulin
- Nephrology and Transplantation Department, Strasbourg University Hospital, Strasbourg, France
| | - Laura Braun-Parvez
- Nephrology and Transplantation Department, Strasbourg University Hospital, Strasbourg, France
| | - Antoine Durrbach
- Department of Nephrology and Transplantation, Bicetre Hospital, APHP, INSERM UMR 1186, Paris-Saclay University, Paris, France
| | - Anne-Elisabeth Heng
- Nephrology, Dialysis and Transplantation Department, Gabriel Montpied Hospital, Clermont-Ferrand, France
| | - Philippe Grimbert
- Department of Nephrology and Transplantation, Henri-Mondor Hospital, APHP, Créteil, France
| | - Didier Ducloux
- Department of Nephrology, Dialysis, and Renal Transplantation, Besançon University Hospital, Besançon, France
| | - Gilles Blancho
- Institut de Transplantation Urologie Néphrologie, Nantes University Hospital, Nantes, France
| | - Pierre Merville
- Department of Nephrology, Transplantation, Dialysis et Apheresis, Bordeaux University Hospital, Bordeaux, France
| | - Gabriel Choukroun
- Department of Nephrology, Internal Medicine, Transplantation, Amiens University Hospital, Amiens, France
| | - Yannick Le Meur
- Department of Nephrology, Brest University Hospital, UMR1227, Brest, France
| | - Cécile Vigneau
- Department of Nephrology, Pontchaillou University Hospital, Rennes, France
| | - Christophe Mariat
- Nephrology, Dialysis and Renal Transplantation Department, Hôpital Nord, Saint-Etienne, France
| | - Lionel Rostaing
- Department of Nephrology, Dialysis, and Organ Transplantation, CHU Rangueil, Toulouse University Hospital, Toulouse, France
| | - Jean-François Subra
- Department of Nephrology, Dialysis and Transplantation, University Hospital, Angers and Centre de Recherche en Cancérologie et Immunologie Nantes-Angers, INSERM, Nantes University, Angers University, Angers, France
| | - Jean-Luc Taupin
- Regional Histocompatibility Laboratory, Saint Louis Hospital, AP-HP, Paris, France
| | - Gérard Lambeau
- Institute of Molecular and Cellular Pharmacology, National Center for Scientific Research, University Côte d’Azur, UMR7275, Valbonne Sophia Antipolis, France
| | - Vincent Esnault
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, Centre Hospitier Universitaire de Nice, Nice, France
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Pasteur 2, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Antoine Sicard
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Pasteur 2, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Barbara Seitz-Polski
- Centre de Référence Maladies Rares Syndrome Néphrotique Idiopathique, Centre Hospitier Universitaire de Nice, Nice, France
- Unité de Recherche Clinique Côte d'Azur (UR2CA), Université Côte d'Azur, Nice, France
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Pasteur 2, Centre Hospitalier Universitaire de Nice, Nice, France
| |
Collapse
|
3
|
Wearne N, Botha F, Manning K, Price B, Barday Z, Post FA, Freercks R, Bertels L, Mtingi-Nkonzombi L, Muller E. Clinical and Histopathological Findings in HIV-positive to HIV-positive Kidney Transplant Recipients. Transplantation 2024:00007890-990000000-00938. [PMID: 39590920 DOI: 10.1097/tp.0000000000005271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
BACKGROUND The spectrum of histological findings in transplanted kidneys from HIV-positive donors to HIV-positive recipients is relatively unexplored. This study describes the type and timing of histological diagnoses observed in this unique cohort. METHODS Adequate biopsies were analyzed at implantation and posttransplant between September 2008 and May 2022. Histological disease spectrum, distributions over time, and relevant clinical characteristics and management were reported for both for-cause and protocol biopsies. RESULTS Twenty-four implantation biopsies from 31 deceased donors and 179 allograft biopsies (100 for-cause, 79 protocol) from 50 recipients were analyzed. Most rejection episodes occurred in the first year posttransplant. Eighteen recipients (36%) had at least 1 episode of biopsy-confirmed acute/chronic T cell-mediated rejection (TCMR) or active antibody-mediated rejection (AMR). Protocol biopsies showed no active AMR or acute/chronic TCMR. However, 9 of 79 biopsies identified borderline/suspicious TCMR. Common nonrejection diagnoses were interstitial fibrosis and tubular atrophy, ascending pyelonephritis, and calcineurin inhibitor toxicity. Classic and suspected HIV-associated nephropathy (HIVAN) were identified in 3 and 6 patients, respectively. Protocol biopsies diagnosed 1 case of classic HIVAN and 6 cases of suspected HIVAN. AMR most adversely affected kidney function and significantly contributed to graft failure. CONCLUSIONS The histological findings in this cohort of HIV-positive kidney transplant recipients who received grafts from unmatched HIV-positive donors revealed a spectrum of abnormalities. Protocol biopsies added to surveillance on borderline rejection and assisted in the recognition of HIVAN. Confirmed rejection occurred in 18 recipients (36%). Understanding the factors contributing to this may assist in the optimization of immunosuppressive protocols in the future.
Collapse
Affiliation(s)
- Nicola Wearne
- Division of Nephrology and Hypertension, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Francois Botha
- Pathcare Laboratories, George, South Africa
- Division of Anatomical Pathology, National Health Laboratory Service/University of Cape Town, Faculty of Health Sciences, Groote Schuur Hospital, Cape Town, South Africa
| | - Kathryn Manning
- Department of Surgery, University of Cape Town, Faculty of Health Sciences, Groote Schuur Hospital, Cape Town, South Africa
| | - Brendon Price
- Division of Anatomical Pathology, National Health Laboratory Service/University of Cape Town, Faculty of Health Sciences, Groote Schuur Hospital, Cape Town, South Africa
| | - Zunaid Barday
- Division of Nephrology and Hypertension, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Frank A Post
- Division/HIV Medicine and Infectious Diseases, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Division/HIV Medicine and Infectious Diseases, King's College London, London, United Kingdom
| | - Robert Freercks
- Department of Medicine, Nelson Mandela University, Livingstone Hospital, Gqeberha, South Africa
| | - Laurie Bertels
- Department of Surgery, University of Cape Town, Faculty of Health Sciences, Groote Schuur Hospital, Cape Town, South Africa
| | | | - Elmi Muller
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
4
|
Udomkarnjananun S, Schagen MR, Hesselink DA. A review of landmark studies on maintenance immunosuppressive regimens in kidney transplantation. ASIAN BIOMED 2024; 18:92-108. [PMID: 39175954 PMCID: PMC11338012 DOI: 10.2478/abm-2024-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Immunosuppressive medications play a pivotal role in kidney transplantation, and the calcineurin inhibitors (CNIs), including cyclosporine A (CsA) and tacrolimus (TAC), are considered as the backbone of maintenance immunosuppressive regimens. Since the introduction of CNIs in kidney transplantation, the incidence of acute rejection has decreased, and allograft survival has improved significantly. However, CNI nephrotoxicity has been a major concern, believed to heavily impact long-term allograft survival and function. To address this concern, several CNI-sparing regimens were developed and studied in randomized, controlled, clinical trials, aiming to reduce CNI exposure and preserve long-term allograft function. However, more recent information has revealed that CNI nephrotoxicity is not the primary cause of late allograft failure, and its histopathology is neither specific nor pathognomonic. In this review, we discuss the historical development of maintenance immunosuppressive regimens in kidney transplantation, covering the early era of transplantation, the CNI-sparing era, and the current era where the alloimmune response, rather than CNI nephrotoxicity, appears to be the major contributor to late allograft failure. Our goal is to provide a chronological overview of the development of maintenance immunosuppressive regimens and summarize the most recent information for clinicians caring for kidney transplant recipients (KTRs).
Collapse
Affiliation(s)
- Suwasin Udomkarnjananun
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
- Excellence Center for Solid Organ Transplantation, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
- Renal Immunology and Transplantation Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok10330, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok10330, Thailand
| | - Maaike R. Schagen
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam3000, The Netherlands
| | - Dennis A. Hesselink
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam3000, The Netherlands
| |
Collapse
|
5
|
Padayachee S, Adam A, Fabian J. The impact of diabetes and hypertension on renal allograft survival- A single center study. Curr Urol 2023; 17:286-291. [PMID: 37994332 PMCID: PMC10662914 DOI: 10.1097/cu9.0000000000000068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/10/2021] [Indexed: 11/26/2022] Open
Abstract
Background To determine the impact of pre-transplant diabetes mellitus (DM) and post-transplant hypertension (HT) at 1 year on renal allograft survival in all adult first kidney-only (FKO) transplant recipients at a single transplant center in Johannesburg, South Africa. Materials and methods A retrospective review was conducted of all adult FKO transplant procedures at the Charlotte Maxeke Johannesburg Academic Hospital transplant unit between 1966 and 2013. Results During the stipulated timeframe, 1685 adult FKO transplant procedures were performed. Of these, 84.1% were from deceased donors (n = 1413/1685). The prevalence of pre-transplant DM transplant recipients with no missing or incomplete records was 6.5% (n = 107/1625). Of the total cohort of 1685 adult FKO transplant recipients, 63.6% of those with no missing data survived to 1 year (n = 1072/1685). The prevalence of HT at 1-year post-transplant was 53.6% (n = 503/1072). HT at 1-year post-transplant, even after adjusted survival analysis, proved a significant risk factor for renal allograft loss (hazard ratio, 1.63; 95% confidence interval, 1.37-1.94) (p < 0.0001). Similarly, after adjusted survival analysis, the risk of renal allograft loss within the pre-transplant DM group was significantly higher (p = 0.043; hazard ratio, 1.26; 95% confidence interval, 1.01-1.58). Conclusions This study identified pre-transplantation diabetes mellitus and post-transplantation HT as significant risk factors for graft loss within the population assessed in this region of the world. These factors could potentially be used as independent predictors of renal graft survival.
Collapse
Affiliation(s)
- Sumesh Padayachee
- Division of Urology, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ahmed Adam
- Division of Urology, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Departments of Urology, Charlotte Maxeke Johannesburg Academic Hospital (CMJAH), Helen Joseph Hospital (HJH), and Rahima; Moosa Mother & Child Hospital (RMMCh); Wits Donald Gordon Medical Center, Johannesburg, South Africa
| | - June Fabian
- Wits Donald Gordon Medical Center, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
6
|
Cleenders E, Koshy P, Van Loon E, Lagrou K, Beuselinck K, Andrei G, Crespo M, De Vusser K, Kuypers D, Lerut E, Mertens K, Mineeva-Sangwo O, Randhawa P, Senev A, Snoeck R, Sprangers B, Tinel C, Van Craenenbroeck A, van den Brand J, Van Ranst M, Verbeke G, Coemans M, Naesens M. An observational cohort study of histological screening for BK polyomavirus nephropathy following viral replication in plasma. Kidney Int 2023; 104:1018-1034. [PMID: 37598855 DOI: 10.1016/j.kint.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 08/22/2023]
Abstract
Systematic screening for BKPyV-DNAemia has been advocated to aid prevention and treatment of polyomavirus associated nephropathy (PyVAN), an important cause of kidney graft failure. The added value of performing a biopsy at time of BKPyV-DNAemia, to distinguish presumptive PyVAN (negative SV40 immunohistochemistry) and proven PyVAN (positive SV40) has not been established. Therefore, we studied an unselected cohort of 950 transplantations, performed between 2008-2017. BKPyV-DNAemia was detected in 250 (26.3%) transplant recipients, and positive SV40 in 91 cases (9.6%). Among 209 patients with a concurrent biopsy at time of first BKPyV-DNAemia, 60 (28.7%) biopsies were SV40 positive. Plasma viral load showed high diagnostic value for concurrent SV40 positivity (ROC-AUC 0.950, 95% confidence interval 0.916-0.978) and the semiquantitatively scored percentage of tubules with evidence of polyomavirus replication (pvl score) (0.979, 0.968-0.988). SV40 positivity was highly unlikely when plasma viral load is below 4 log10 copies/ml (negative predictive value 0.989, 0.979-0.994). In SV40 positive patients, higher plasma BKPyV-DNA load and higher pvl scores were associated with slower viral clearance from the blood (hazard ratio 0.712, 95% confidence interval 0.604-0.839, and 0.327, 0.161-0.668, respectively), whereas the dichotomy positivity/negativity of SV40 immunohistochemistry did not predict viral clearance. Although the pvl score offers some prognostic value for viral clearance on top of plasma viral load, the latter provided good guidance for when a biopsy was unnecessary to exclude PyVAN. Thus, the distinction between presumptive and proven PyVAN, based on SV40 immunohistochemistry, has limited clinical value. Hence, management of BKPyV-DNAemia and immunosuppression reduction should be weighed against the risk of occurrence of rejection, or exacerbation of rejection observed concomitantly.
Collapse
Affiliation(s)
- Evert Cleenders
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Public Health and Primary Care, Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, Belgium
| | - Priyanka Koshy
- Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium
| | - Kurt Beuselinck
- Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Graciela Andrei
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar Medical Research Institute (IMIM), Hospital del Mar, Barcelona, Spain
| | - Katrien De Vusser
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Dirk Kuypers
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Evelyne Lerut
- Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Kris Mertens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Olga Mineeva-Sangwo
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Parmjeet Randhawa
- Division of Transplantation Pathology, the Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center-Montefiore Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aleksandar Senev
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Robert Snoeck
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Molecular Immunology, KU Leuven, Leuven, Belgium
| | - Claire Tinel
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Amaryllis Van Craenenbroeck
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Jan van den Brand
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Marc Van Ranst
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Geert Verbeke
- Department of Public Health and Primary Care, Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, Belgium
| | - Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Public Health and Primary Care, Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium; Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Redondo-Pachón D, Calatayud E, Buxeda A, Pérez-Sáez MJ, Arias-Cabrales C, Gimeno J, Burballa C, Mir M, Llinàs-Mallol L, Outon S, Pascual J, Crespo M. Evolution of kidney allograft loss causes over 40 years (1979-2019). Nefrologia 2023; 43:316-327. [PMID: 37507293 DOI: 10.1016/j.nefroe.2023.07.003] [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: 07/23/2021] [Accepted: 12/01/2021] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION The improvement of kidney allograft recipient and graft survival showed a decrease over the last 40 years. Long-term graft loss rate remained stable during a 25-year time span. Knowing the changing causes and the risk factors associated with graft loss requires special attention. The present study aimed to assess the causes of graft loss and kidney allograft recipient death. Also, we aimed to compare two different periods (1979-1999 and 2000-2019) to identify changes in the characteristics of the failed allografts and recipient and donors profile. METHODS AND PATIENTS We performed a single-center cohort study. We included all the kidney transplant recipients at the Hospital del Mar (Barcelona) between May 1979 and December 2019. Graft loss was defined as recipient death with functioning graft and as loss of graft function (return to dialysis or retransplantation). We assessed the causes of graft loss using clinical and histological information. We also analyzed the results of the two different transplant periods (1979-1999 and 2000-2019). RESULTS Between 1979 and 2019, 1522 transplants were performed. The median follow-up time was 56 (IQR 8-123) months. During follow-up, 722 (47.5%) grafts were lost: 483 (66.9%) due to graft failure and 239 (33.1%) due to death with functioning graft. The main causes of death were cardiovascular (25.1%), neoplasms (25.1%), and infectious diseases (21.8%). These causes were stable between the two periods of time. Only the unknown cause of death has decreased in the last period. The main cause of graft failure (loss of graft function) was the allograft chronic dysfunction (75%). When histologic information was available, antibody-mediated rejection (ABMR) and interstitial fibrosis/tubular atrophy (IF/TA) were the most frequent specific causes (15.9% and 12.6%). Of the graft failures, 213 (29.5%) were early (<1 year of transplantation). Vascular thrombosis was the main cause of early graft failure in the second period (2000-2019) (46.7%) and T-cell-mediated rejection (TCMR) was the main cause (31.3%) in the first period (1979-1999). The causes of late graft loss were similar between the two periods. CONCLUSIONS The causes of kidney allograft recipient death are still due to cardiovascular and malignant diseases. Vascular thrombosis has emerged as a frequent cause of early graft loss in the most recent years. The evaluation of the causes of graft loss is necessary to improve kidney transplantation outcomes.
Collapse
Affiliation(s)
| | - Emma Calatayud
- Servicio de Nefrología, Hospital Universitario Doctor Peset, Valencia, Spain
| | - Anna Buxeda
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| | | | | | - Javier Gimeno
- Servicio de Anatomía Patológica, Hospital del Mar, Barcelona, Spain
| | - Carla Burballa
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| | - Marisa Mir
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| | | | - Sara Outon
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| | - Julio Pascual
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| | - Marta Crespo
- Servicio de Nefrología, Hospital del Mar, Barcelona, Spain
| |
Collapse
|
8
|
Seeking Standardized Definitions for HLA-incompatible Kidney Transplants: A Systematic Review. Transplantation 2023; 107:231-253. [PMID: 35915547 DOI: 10.1097/tp.0000000000004262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is no standard definition for "HLA incompatible" transplants. For the first time, we systematically assessed how HLA incompatibility was defined in contemporary peer-reviewed publications and its prognostic implication to transplant outcomes. METHODS We combined 2 independent searches of MEDLINE, EMBASE, and the Cochrane Library from 2015 to 2019. Content-expert reviewers screened for original research on outcomes of HLA-incompatible transplants (defined as allele or molecular mismatch and solid-phase or cell-based assays). We ascertained the completeness of reporting on a predefined set of variables assessing HLA incompatibility, therapies, and outcomes. Given significant heterogeneity, we conducted narrative synthesis and assessed risk of bias in studies examining the association between death-censored graft failure and HLA incompatibility. RESULTS Of 6656 screened articles, 163 evaluated transplant outcomes by HLA incompatibility. Most articles reported on cytotoxic/flow T-cell crossmatches (n = 98). Molecular genotypes were reported for selected loci at the allele-group level. Sixteen articles reported on epitope compatibility. Pretransplant donor-specific HLA antibodies were often considered (n = 143); yet there was heterogeneity in sample handling, assay procedure, and incomplete reporting on donor-specific HLA antibodies assignment. Induction (n = 129) and maintenance immunosuppression (n = 140) were frequently mentioned but less so rejection treatment (n = 72) and desensitization (n = 70). Studies assessing death-censored graft failure risk by HLA incompatibility were vulnerable to bias in the participant, predictor, and analysis domains. CONCLUSIONS Optimization of transplant outcomes and personalized care depends on accurate HLA compatibility assessment. Reporting on a standard set of variables will help assess generalizability of research, allow knowledge synthesis, and facilitate international collaboration in clinical trials.
Collapse
|
9
|
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: 17] [Impact Index Per Article: 5.7] [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
|
10
|
Integrated Immunologic Monitoring in Solid Organ Transplantation: The Road Toward Torque Teno Virus-guided Immunosuppression. Transplantation 2022; 106:1940-1951. [PMID: 35509090 PMCID: PMC9521587 DOI: 10.1097/tp.0000000000004153] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Potent immunosuppressive drugs have been introduced into clinical care for solid organ transplant recipients. It is now time to guide these drugs on an individual level to optimize their efficacy. An ideal tool simultaneously detects overimmunosuppression and underimmunosuppression, is highly standardized, and is straightforward to implement into routine. Randomized controlled interventional trials are crucial to demonstrate clinical value. To date, proposed assays have mainly focused on the prediction of rejection and were based on the assessment of few immune compartments. Recently, novel tools have been introduced based on a more integrated approach to characterize the immune function and cover a broader spectrum of the immune system. In this respect, the quantification of the plasma load of a highly prevalent and apathogenic virus that might reflect the immune function of its host has been proposed: the torque teno virus (TTV). Although TTV control is driven by T cells, other major immune compartments might contribute to the hosts' response. A standardized in-house polymerase chain reaction and a conformité européenne-certified commercially available polymerase chain reaction are available for TTV quantification. TTV load is associated with rejection and infection in solid organ transplant recipients, and cutoff values for risk stratification of such events have been proposed for lung and kidney transplantation. Test performance of TTV load does not allow for the diagnosis of rejection and infection but is able to define at-risk patients. Hitherto TTV load has not been used in interventional settings, but two interventional randomized controlled trials are currently testing the safety and efficacy of TTV-guided immunosuppression.
Collapse
|
11
|
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. The natural history of de novo donor-specific HLA antibodies after kidney transplantation. Front Med (Lausanne) 2022; 9:943502. [PMID: 36186822 PMCID: PMC9523126 DOI: 10.3389/fmed.2022.943502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022] Open
Abstract
Background De novo donor-specific HLA antibodies (dnDSA) are key factors in the diagnosis of antibody-mediated rejection (ABMR) and related to graft loss. Methods This retrospective study was designed to evaluate the natural course of dnDSA in graft function and kidney allograft survival and to assess the impact of mean fluorescence intensity (MFI) evolution as detected by annual Luminex® screening. All 400 kidney transplant recipients with 731 dnDSA against the last graft (01/03/2000-31/05/2021) were included. Results During 8.3 years of follow-up, ABMR occurred in 24.8% and graft loss in 33.3% of the cases, especially in patients with class I and II dnDSA, and those with multiple dnDSA. We observed frequent changes in MFI with 5-year allograft survivals post-dnDSA of 74.0% in patients with MFI reduction ≥ 50%, 62.4% with fluctuating MFI (MFI reduction ≥ 50% and doubling), and 52.7% with doubling MFI (log-rank p < 0.001). Interestingly, dnDSA in 168 (24.3%) cases became negative at some point during follow-up, and 38/400 (9.5%) patients became stable negative, which was associated with better graft survival. Multivariable analysis revealed the importance of MFI evolution and rejection, while class and number of dnDSA were not contributors in this model. Conclusion In summary, we provide an in-depth analysis of the natural course of dnDSA after kidney transplantation, first evidence for the impact of MFI evolution on graft outcomes, and describe a relevant number of patients with a stable disappearance of dnDSA, related to better allograft survival.
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
- Valdecilla Biomedical Research Institute (IDIVAL), Santander, Spain
- *Correspondence: Covadonga López del Moral,
| | - 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
|
12
|
Roufosse C, Becker JU, Rabant M, Seron D, Bellini MI, Böhmig GA, Budde K, Diekmann F, Glotz D, Hilbrands L, Loupy A, Oberbauer R, Pengel L, Schneeberger S, Naesens M. Proposed Definitions of Antibody-Mediated Rejection for Use as a Clinical Trial Endpoint in Kidney Transplantation. Transpl Int 2022; 35:10140. [PMID: 35669973 PMCID: PMC9163810 DOI: 10.3389/ti.2022.10140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022]
Abstract
Antibody-mediated rejection (AMR) is caused by antibodies that recognize donor human leukocyte antigen (HLA) or other targets. As knowledge of AMR pathophysiology has increased, a combination of factors is necessary to confirm the diagnosis and phenotype. However, frequent modifications to the AMR definition have made it difficult to compare data and evaluate associations between AMR and graft outcome. The present paper was developed following a Broad Scientific Advice request from the European Society for Organ Transplantation (ESOT) to the European Medicines Agency (EMA), which explored whether updating guidelines on clinical trial endpoints would encourage innovations in kidney transplantation research. ESOT considers that an AMR diagnosis must be based on a combination of histopathological factors and presence of donor-specific HLA antibodies in the recipient. Evidence for associations between individual features of AMR and impaired graft outcome is noted for microvascular inflammation scores ≥2 and glomerular basement membrane splitting of >10% of the entire tuft in the most severely affected glomerulus. Together, these should form the basis for AMR-related endpoints in clinical trials of kidney transplantation, although modifications and restrictions to the Banff diagnostic definition of AMR are proposed for this purpose. The EMA provided recommendations based on this Broad Scientific Advice request in December 2020; further discussion, and consensus on the restricted definition of the AMR endpoint, is required.
Collapse
Affiliation(s)
- Candice Roufosse
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, London, United Kingdom
| | - Jan Ulrich Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Marion Rabant
- Department of Pathology, Hôpital Necker-Enfants Malades, Paris, France
| | - Daniel Seron
- Department of Nephrology and Kidney Transplantation, Vall d'Hebrón University Hospital, Barcelona, Spain
| | | | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Internal Medicine, Medical University of Vienna, Vienna, Austria
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Fritz Diekmann
- Department of Nephrology and Kidney Transplantation, Hospital Clinic Barcelona, Barcelona, Spain
| | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, Hôpital Saint Louis, Paris, France
| | - Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Internal Medicine, Medical University of Vienna, Vienna, Austria
| | - Liset Pengel
- Centre for Evidence in Transplantation, University of Oxford, Oxford, United Kingdom
| | - 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
| |
Collapse
|
13
|
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
|
14
|
Wu K, Schmidt D, López del Moral C, Osmanodja B, Lachmann N, Halleck F, Choi M, Bachmann F, Ronicke S, Duettmann W, Naik M, Schrezenmeier E, Rudolph B, Budde K. Poor Outcomes in Patients With Transplant Glomerulopathy Independent of Banff Categorization or Therapeutic Interventions. Front Med (Lausanne) 2022; 9:889648. [PMID: 35646957 PMCID: PMC9133540 DOI: 10.3389/fmed.2022.889648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTransplant glomerulopathy (TG) may indicate different disease entities including chronic AMR (antibody-mediated rejection). However, AMR criteria have been frequently changed, and long-term outcomes of allografts with AMR and TG according to Banff 2017 have rarely been investigated.Methods282 kidney allograft recipients with biopsy-proven TG were retrospectively investigated and diagnosed according to Banff'17 criteria: chronic AMR (cAMR, n = 72), chronic active AMR (cAAMR, n = 76) and isolated TG (iTG, n = 134). Of which 25/72 (34.7%) patients of cAMR group and 46/76 (60.5%) of cAAMR group were treated with antihumoral therapy (AHT).ResultsUp to 5 years after indication biopsy, no statistically significant differences were detected among iTG, cAMR and cAAMR groups in annual eGFR decline (−3.0 vs. −2.0 vs. −2.8 ml/min/1.73 m2 per year), 5-year median eGFR (21.5 vs. 16.0 vs. 20.0 ml/min/1.73 m2), 5-year graft survival rates (34.1 vs. 40.6 vs. 31.8%) as well as urinary protein excretion during follow-up. In addition, cAMR and cAAMR patients treated with AHT had similar graft and patient survival rates in comparison with those free of AHT, and similar comparing with iTG group. The TG scores were not associated with 5-year postbiopsy graft failure; whereas the patients with higher scores of chronic allograft scarring (by mm-, ci- and ct-lesions) had significantly lower graft survival rates than those with mild scores. The logistic-regression analysis demonstrated that Banff mm-, ah-, t-, ci-, ct-lesions and the eGFR level at biopsy were associated with 5-year graft failure.ConclusionsThe occurrence of TG is closely associated with graft failure independent of disease categories and TG score, and the long-term clinical outcomes were not influenced by AHT. The Banff lesions indicating progressive scarring might be better suited to predict an unfavorable outcome.
Collapse
Affiliation(s)
- Kaiyin Wu
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
- *Correspondence: Kaiyin Wu
| | - Danilo Schmidt
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Covadonga López del Moral
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Lachmann
- HLA Laboratory, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, BIH, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Ronicke
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wiebke Duettmann
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcel Naik
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| | - Birgit Rudolph
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universitätzu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin Institute of Health (BIH), Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
15
|
Death With Function and Graft Failure After Kidney Transplantation: Risk Factors at Baseline Suggest New Approaches to Management. Transplant Direct 2022; 8:e1273. [PMID: 35047660 PMCID: PMC8759617 DOI: 10.1097/txd.0000000000001273] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/27/2021] [Indexed: 11/26/2022] Open
Abstract
Background Improving both patient and graft survival after kidney transplantation are major unmet needs. The goal of this study was to assess risk factors for specific causes of graft loss to determine to what extent patients who develop either death with a functioning graft (DWFG) or graft failure (GF) have similar baseline risk factors for graft loss. Methods We retrospectively studied all solitary renal transplants performed between January 1, 2006, and December 31, 2018, at 3 centers and determined the specific causes of DWFG and GF. We examined outcomes in different subgroups using competing risk estimates and cause-specific Cox models. Results Of the 5752 kidney transplants, graft loss occurred in 21.6% (1244) patients, including 12.0% (691) DWFG and 9.6% (553) GF. DWFG was most commonly due to malignancy (20.0%), infection (19.7%), cardiac disease (12.6%) with risk factors of older age and pretransplant dialysis, and diabetes as the cause of renal failure. For GF, alloimmunity (38.7%), glomerular diseases (18.6%), and tubular injury (13.9%) were the major causes. Competing risk incidence models identified diabetes and older recipients with higher rates of both DWFG and nonalloimmune GF. Conclusions These data suggest that at baseline, 2 distinct populations can be identified who are at high risk for renal allograft loss: a younger, nondiabetic patient group who develops GF due to alloimmunity and an older, more commonly diabetic population who develops DWFG and GF due to a mixture of causes-many nonalloimmune. Individualized management is needed to improve long-term renal allograft survival in the latter group.
Collapse
|
16
|
Redondo-Pachón D, Calatayud E, Buxeda A, Pérez-Sáez MJ, Arias-Cabrales C, Gimeno J, Burballa C, Mir M, Llinàs-Mallol L, Outon S, Pascual J, Crespo M. Evolución de las causas de pérdida del injerto en trasplante renal durante 40 años (1979-2019). Nefrologia 2021. [DOI: 10.1016/j.nefro.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
17
|
Doberer K, Haupenthal F, Nackenhorst M, Bauernfeind F, Dermuth F, Eigenschink M, Schiemann M, Kläger J, Görzer I, Eskandary F, Reindl-Schwaighofer R, Kikić Ž, Böhmig G, Strassl R, Regele H, Puchhammer-Stöckl E, Bond G. Torque Teno Virus Load Is Associated With Subclinical Alloreactivity in Kidney Transplant Recipients: A Prospective Observational Trial. Transplantation 2021; 105:2112-2118. [PMID: 33587432 PMCID: PMC8376270 DOI: 10.1097/tp.0000000000003619] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nonpathogenic torque teno viruses (TTVs) are highly prevalent in transplant recipients and associated with immunosuppression. Studies in kidney transplant patients have proposed assessment of TTV load for risk stratification of clinically overt graft rejection. The value of TTV quantification in the context of subclinical rejection has not been evaluated. METHODS In this prospective trial, 307 consecutive kidney transplant recipients were subjected to per-protocol monitoring of plasma TTV. TTV was analyzed in the context of protocol biopsies (n = 82), scheduled 1 year posttransplantation. RESULTS TTV load at the time of biopsy was lower in recipients with rejection (n = 19; according to Banff, including borderline changes suspicious for acute T cell-mediated rejection) than those without rejection (n = 63) whereby each log increase in TTV copies/mL decreased the risk for rejection by 9% (risk ratio 0.91, 95% confidence interval, 0.85-0.97; P = 0.004). Development of chronic lesions (cg, cv, ci, ct, ah, ptcml) was associated with the number of days with a TTV load <1 × 106 copies/mL between months 3 and 12 posttransplant (β 0.07, 95% confidence interval, 0.01-0.14; P = 0.02). CONCLUSIONS This trial demonstrates an association between TTV and subclinical graft rejection in kidney transplant recipients. A TTV load <1 × 106 copies/mL suggests suboptimal immunosuppression.
Collapse
Affiliation(s)
- Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Frederik Haupenthal
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Maja Nackenhorst
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Florian Bauernfeind
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florentina Dermuth
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Eigenschink
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Martin Schiemann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Johannes Kläger
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Irene Görzer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Željko Kikić
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Georg Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Robert Strassl
- Division of Clinical Virology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Heinz Regele
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | | | - Gregor Bond
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Mayrdorfer M, Liefeldt L, Wu K, Rudolph B, Zhang Q, Friedersdorff F, Lachmann N, Schmidt D, Osmanodja B, Naik MG, Duettmann W, Halleck F, Merkel M, Schrezenmeier E, Waiser J, Duerr M, Budde K. Exploring the Complexity of Death-Censored Kidney Allograft Failure. J Am Soc Nephrol 2021; 32:1513-1526. [PMID: 33883251 PMCID: PMC8259637 DOI: 10.1681/asn.2020081215] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/04/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Few studies have thoroughly investigated the causes of kidney graft loss (GL), despite its importance. METHODS A novel approach assigns each persistent and relevant decline in renal function over the lifetime of a renal allograft to a standardized category, hypothesizing that singular or multiple events finally lead to GL. An adjudication committee of three physicians retrospectively evaluated indication biopsies, laboratory testing, and medical history of all 303 GLs among all 1642 recipients of transplants between January 1, 1997 and December 31, 2017 at a large university hospital to assign primary and/or secondary causes of GL. RESULTS In 51.2% of the patients, more than one cause contributed to GL. The most frequent primary or secondary causes leading to graft failure were intercurrent medical events in 36.3% of graft failures followed by T cell-mediated rejection (TCMR) in 34% and antibody-mediated rejection (ABMR) in 30.7%. In 77.9%, a primary cause could be attributed to GL, of which ABMR was most frequent (21.5%). Many causes for GL were identified, and predominant causes for GL varied over time. CONCLUSIONS GL is often multifactorial and more complex than previously thought.
Collapse
Affiliation(s)
- Manuel Mayrdorfer
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lutz Liefeldt
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kaiyin Wu
- Department of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Rudolph
- Department of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Qiang Zhang
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Nils Lachmann
- Institute for Transfusion Medicine, HLA Laboratory, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel G. Naik
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany,BIH, Berlin Institute of Health, Berlin, Germany
| | - Wiebke Duettmann
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Marina Merkel
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany,BIH, Berlin Institute of Health, Berlin, Germany
| | - Johannes Waiser
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Duerr
- 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
| |
Collapse
|
19
|
Groeneweg KE, van der Toorn FA, Roelen DL, van Kooten C, Heidt S, Claas FHJ, Reinders MEJ, de Fijter JW, Soonawala D. Single antigen testing to reduce early antibody-mediated rejection risk in female recipients of a spousal donor kidney. Transpl Immunol 2021; 67:101407. [PMID: 33975014 DOI: 10.1016/j.trim.2021.101407] [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: 12/04/2020] [Revised: 04/25/2021] [Accepted: 05/06/2021] [Indexed: 11/19/2022]
Abstract
Female recipients of a spousal donor kidney transplant are at greater risk of donor-specific pre-immunization, which may increase the risk of acute antibody-mediated rejection (ABMR). We assessed the incidence of early ABMR (within two weeks after transplantation), risk factors for ABMR and graft function in 352 complement-dependent cytotoxicity test-negative LURD transplant recipients, transplanted between 1997 and 2014 at the Leiden University Medical Center in The Netherlands. Risk factors for immunization were retrieved from the health records. As methods to screen for preformed donor-specific antibodies (pDSA) have developed through time, we retrospectively screened those with ABMR for pDSA using pooled-antigen bead (PAB) and single-antigen bead (SAB) assays. The cumulative incidence of rejection in the first six months after transplantation was 18% (TCMR 15%; early ABMR 3%). Early ABMR resulted in inferior graft survival and was more common in women who received a kidney from their spouse (10%) than in other women (2%) and men (<1%). The SAB assay retrospectively identified pDSA in seven of nine cases of early ABMR (78%), while the PAB detected pDSA in only three cases (33%). Seeing that early ABMR occurred in 10% of women who received a kidney from their spouse, a SAB assay should be included in the pre-transplant assessment of this group of women, regardless of the result of the PAB assay.
Collapse
Affiliation(s)
- Koen E Groeneweg
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, the Netherlands.
| | | | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, the Netherlands
| | - Cees van Kooten
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, the Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, the Netherlands
| | - Frans H J Claas
- Department of Immunology, Leiden University Medical Center, the Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, the Netherlands
| | - Johan W de Fijter
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, the Netherlands
| | - Darius Soonawala
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, the Netherlands; Department of Internal Medicine, Haga Teaching Hospital, The Hague, the Netherlands
| |
Collapse
|
20
|
Naciri Bennani H, Daligault M, Noble J, Bardy B, Motte L, Giovannini D, Emprou C, Fiard G, Imerzoukene F, Bourdin A, Masson D, Janbon B, Malvezzi P, Rostaing L, Jouve T. Treatment of antibody-mediated rejection with double-filtration plasmapheresis, low dose IVIg plus rituximab after kidney transplantation. J Clin Apher 2021; 36:584-594. [PMID: 33783868 DOI: 10.1002/jca.21897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/16/2021] [Indexed: 12/19/2022]
Abstract
Antibody-mediated rejection (ABMR) at early or late post-transplantation remains challenging. We performed a single-center single-arm study where four cases of acute ABMR and nine cases of chronic active ABMR (defined by Banff classification) were treated with double-filtration plasmapheresis (two cycles of three consecutive daily sessions with a 4-day gap between). At the end of the third and sixth DFPP sessions, the patients received rituximab 375 mg/m2 . After a median follow-up of 1078 (61-1676) days, kidney-allograft survival was 50%. Before DFPP/rituximab therapy, the median donor-specific alloantibody (DSA) mean fluorescence intensity (MFI) was 9160 (4000-15 400); 45 days (D45) later it had significantly decreased to 7375 (215-18 100) (P = .018). In addition, at one-year (Y1) post-therapy, MFI had decreased further, that is, 4060 (400-7850) (P = .001). In two patients, DSA MFIs decreased and remained below 2000. The slope of estimated glomerular-filtration rate within the 6 months preceding intervention was -1.18 mL/min/month and remained unchanged at -1.29 mL/min/month within the year after intervention. Proteinuria remained unchanged. Baseline Banff scores on repeat allograft biopsies (post-therapy D45, Y1) did not show any improvement. Side-effects were mild to moderate. We conclude that the combined DFPP/rituximab significantly decreased DSAs in ABMR kidney-transplant recipients but did not improve renal function or renal histology at 1-year follow-up.
Collapse
Affiliation(s)
- Hamza Naciri Bennani
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | - Mélanie Daligault
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | - Johan Noble
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | | | - Lionel Motte
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | - Diane Giovannini
- Pathology Department, Grenoble University Hospital, Grenoble, France
| | - Camille Emprou
- Pathology Department, Grenoble University Hospital, Grenoble, France
| | - Gaëlle Fiard
- Department of Urology and Kidney Transplantation, Grenoble University Hospital, Grenoble, France.,Grenoble Alpes University, Grenoble, France
| | - Farida Imerzoukene
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | | | | | - Bénédicte Janbon
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | - Paolo Malvezzi
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France
| | - Lionel Rostaing
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France.,Grenoble Alpes University, Grenoble, France
| | - Thomas Jouve
- Nephrology, Hemodialysis, Apheresis and Kidney Transplantation Department, Grenoble University Hospital, Grenoble, France.,Grenoble Alpes University, Grenoble, France
| |
Collapse
|
21
|
Cheung D, Garcia J, Beduschi T, Langshaw A, Arheart K, Wunsch C, Vianna R, Gonzalez IA. Re-evaluating Blood Markers as Predictors of Outcome in Multivisceral and Intestinal Transplantation. Transplant Proc 2021; 53:696-704. [PMID: 33558087 DOI: 10.1016/j.transproceed.2021.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/08/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Multivisceral transplant (MVTx) and isolated intestinal transplant (ITx) are complex surgical procedures. The subsequent proinflammatory state in the immediate postoperative period makes interpretation of blood markers difficult. METHOD We aimed to establish the course of various blood markers after MVTx/ITx, and to evaluate their use as diagnostic markers of complications. This was a single center prospective cohort. We analyzed blood markers collected preoperatively, on alternate days for the first postoperative week, and then weekly for 4 weeks. This study was in compliance with The Declaration of Helsinki. RESULTS Over a 16-month period (July 2017-October 2018), 20 subjects aged 2 to 67 years with a median age of 24.5 years received MVTx/ITx. Twelve recipients (60%) had an infection. Neutrophil lymphocyte count ratio (NLCR) was higher than established upper limits of normal, regardless of infection status. NLCR and white blood cell count were useful to identify infected MVTx/ITx recipients, with P values <.05 for 2 and 1 of 7 time points post transplant, respectively. Higher preoperative eosinophil% predicted future acute cellular rejection (P value .023). CONCLUSIONS This is the first study to extensively track the course of blood markers post MVTx/ITx and identified NLCR and white blood cell count as potential diagnostic blood markers of infection.
Collapse
Affiliation(s)
- Donna Cheung
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Miami, Miami, Florida.
| | - Jennifer Garcia
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Miami, Miami, Florida
| | - Thiago Beduschi
- Department of Surgery, Division of Liver/GI Transplant, Miami Transplant Institute, Jackson Memorial Hospital/University of Miami, Miami, Florida
| | - Amber Langshaw
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Miami, Miami, Florida
| | - Kristopher Arheart
- Department of Public Health Sciences, Division of Biostatistics, University of Miami, Miami, Florida
| | - Chris Wunsch
- Department of Pathology, University of Miami, Miami, Florida
| | - Rodrigo Vianna
- Department of Surgery, Division of Liver/GI Transplant, Miami Transplant Institute, Jackson Memorial Hospital/University of Miami, Miami, Florida
| | - Ivan A Gonzalez
- Department of Pediatrics, Division of Pediatric Infectious Disease and Immunology, University of Miami, Miami, Florida
| |
Collapse
|
22
|
Vincenzi P, Gonzalez J, Guerra G, Gaynor JJ, Alvarez A, Ciancio G. Complex Surgical Reconstruction of Upper Pole Artery in Living-Donor Kidney Transplantation. Ann Transplant 2021; 26:e926850. [PMID: 33446626 PMCID: PMC7814512 DOI: 10.12659/aot.926850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background The use of allografts with multiple renal arteries has increased in the era of laparoscopic donor nephrectomy. Although several studies recommend reconstructing lower pole arteries (LPAs) to reduce risk of urologic complications, it is common opinion to ligate upper pole arteries (UPAs) with a diameter less than 2 mm because of increased risk of thrombosis related to their reconstruction. This retrospective study evaluates the feasibility and safety of reconstructing thin UPAs during living-donor kidney transplantation, with the goal of maintaining the integrity of the graft and assuring its maximal function. Material/Methods Data from 922 living-donor kidney transplants performed between 2009 and 2019 were reviewed. Six cases with UPAs were identified (0.65%). The study endpoints were incidence of allograft vascular and urologic complications, slow graft function, delayed graft function, graft failure, and graft and patient survival. Results The UPAs had a mean diameter of 1.8±0.28 mm. Methods of reconstruction included: interposition graft (n=2), end-to-side anastomosis inside the renal hilum to a branch of the main renal artery (n=3), and side-to-side anastomosis with the main renal artery (n=1). Additional reconstruction of LPAs (n=2) and main renal arteries (n=2) was performed. During a median (range) follow-up of 14.5 (9–49) months no complications were observed. Conclusions Ex vivo reconstruction of UPAs with a diameter less than 2 mm is worth attempting, particularly in the setting of living-donor kidney transplantation.
Collapse
Affiliation(s)
- Paolo Vincenzi
- Department of Surgery, Miami Transplant Institute, Miami, FL, USA.,University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Javier Gonzalez
- Department of Urology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Giselle Guerra
- University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA.,Department of Medicine, Division of Nephrology, Miami Transplant Institute, Miami, FL, USA
| | - Jeffrey J Gaynor
- Department of Surgery, Miami Transplant Institute, Miami, FL, USA.,University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA
| | - Angel Alvarez
- Department of Surgery, Miami Transplant Institute, Miami, FL, USA
| | - Gaetano Ciancio
- Department of Surgery, Miami Transplant Institute, Miami, FL, USA.,University of Miami Miller School of Medicine, Jackson Memorial Hospital, Miami, FL, USA.,Department of Urology, Miami Transplant Institute, Miami, FL, USA
| |
Collapse
|
23
|
Chronic Histologic Changes Are Present Regardless of HLA Mismatches: Evidence from HLA Identical Living Donor Kidney Transplants. Transplantation 2020; 105:e244-e256. [PMID: 33315759 DOI: 10.1097/tp.0000000000003579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND At 5 and 10 years after kidney transplantation, chronic histologic changes such as arteriolar hyalinosis and mesangial expansion are common, however, determining etiology is difficult. We compared surveillance biopsies in living donor kidney transplants (LDKTx) from HLA matched siblings (termed HLA-identical (HLA-ID)) to HLA non-ID to investigate which histologic changes were likely due to alloimmune injury and which were due to non-alloimmune injury. METHODS We performed a retrospective, cohort study comparing HLA-ID sibling LDKTx (n=175) to HLA non-ID LDKTx (n=175; matched for age, sex and year of transplant +/- 2 years) performed at a single institution from 03/1999 to 11/2018. RESULTS Baseline characteristics and maintenance immunosuppression were similar. Mortality rates were similar, but in the HLA-ID group, 10-year death-censored graft survival was higher (93.8% vs 80.9% HLA non-ID LDKTx, p<0.001), rejection rates were lower (after 1 year 9.6% vs 27.1%; p<0.001) and Banff inflammation scores including glomerulitis and peritubular capillaritis were lower on surveillance biopsies at 1, 5 and 10 years. In contrast, chronic Banff scores (interstitial fibrosis, arteriolar hyalinosis, mesangial expansion, etc.) were similar in prevalence and severity on surveillance biopsies at 1, 5 and 10 years. CONCLUSIONS HLA-ID LDKTx have less inflammation and less transplant glomerulopathy, but most chronic histologic changes were similar to less-well matched LDKTx. We conclude that these types of chronic changes are not associated with HLA mismatches and may be due to non-immunologic causes (hypertension, obesity, etc.) suggesting that new management approaches to prevent these lesions may be needed.
Collapse
|
24
|
Circulating Long Noncoding RNA LNC-EPHA6 Associates with Acute Rejection after Kidney Transplantation. Int J Mol Sci 2020; 21:ijms21165616. [PMID: 32764470 PMCID: PMC7460577 DOI: 10.3390/ijms21165616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/21/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Acute rejection (AR) of a kidney graft in renal transplant recipients is associated with microvascular injury in graft dysfunction and, ultimately, graft failure. Circulating long noncoding RNAs (lncRNAs) may be suitable markers for vascular injury in the context of AR. Here, we first investigated the effect of AR after kidney transplantation on local vascular integrity and demonstrated that the capillary density markedly decreased in AR kidney biopsies compared to pre-transplant biopsies. Subsequently, we assessed the circulating levels of four lncRNAs (LNC-RPS24, LNC-EPHA6, MALAT1, and LIPCAR), that were previously demonstrated to associate with vascular injury in a cohort of kidney recipients with a stable kidney transplant function (n = 32) and recipients with AR (n = 15). The latter were followed longitudinally six and 12 months after rejection. We found higher levels of circulating LNC-EPHA6 during rejection, compared with renal recipients with a stable kidney function (p = 0.017), that normalized one year after AR. In addition, LNC-RPS24, LNC-EPHA6, and LIPCAR levels correlated significantly with the vascular injury marker soluble thrombomodulin. We conclude that AR and microvascular injury are associated with higher levels of circulating LNC-EPHA6, which emphasizes the potential role of lncRNAs as biomarker in the context of AR.
Collapse
|
25
|
Doberer K, Schiemann M, Strassl R, Haupenthal F, Dermuth F, Görzer I, Eskandary F, Reindl‐Schwaighofer R, Kikić Ž, Puchhammer‐Stöckl E, Böhmig GA, Bond G. Torque teno virus for risk stratification of graft rejection and infection in kidney transplant recipients-A prospective observational trial. Am J Transplant 2020; 20:2081-2090. [PMID: 32034850 PMCID: PMC7496119 DOI: 10.1111/ajt.15810] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/28/2020] [Accepted: 01/31/2020] [Indexed: 01/25/2023]
Abstract
The nonpathogenic and ubiquitous torque teno virus (TTV) is associated with immunosuppression in solid organ transplant recipients. Studies in kidney transplant patients proposed TTV quantification for risk stratification of graft rejection and infection. In this prospective trial (DRKS00012335) 386 consecutive kidney transplant recipients were subjected to longitudinal per-protocol monitoring of plasma TTV load by polymerase chain reaction for 12 months posttransplant. TTV load peaked at the end of month 3 posttransplant and reached steady state thereafter. TTV load after the end of month 3 was analyzed in the context of subsequent rejection diagnosed by indication biopsy and infection within the first year posttransplant, respectively. Each log increase in TTV load decreased the odds for rejection by 22% (odds ratio [OR] 0.78, 95% confidence interval [CI] 0.62-0.97; P = .027) and increased the odds for infection by 11% (OR 1.11, 95% CI 1.06-1.15; P < .001). TTV was quantified at a median of 14 days before rejection was diagnosed and 27 days before onset of infection, respectively. We defined a TTV load between 1 × 106 and 1 × 108 copies/mL as optimal range to minimize the risk for rejection and infection. These data support the initiation of an interventional trial assessing the efficacy of TTV-guided immunosuppression to reduce infection and graft rejection in kidney transplant recipients.
Collapse
Affiliation(s)
- Konstantin Doberer
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | - Martin Schiemann
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | - Robert Strassl
- Division of VirologyDepartment of Laboratory MedicineMedical University ViennaViennaAustria
| | - Frederik Haupenthal
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | - Florentina Dermuth
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | - Irene Görzer
- Center for VirologyMedical University ViennaViennaAustria
| | - Farsad Eskandary
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | | | - Željko Kikić
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | | | - Georg A. Böhmig
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| | - Gregor Bond
- Division of Nephrology and DialysisDepartment of Medicine IIIMedical University ViennaViennaAustria
| |
Collapse
|
26
|
A Review on the Function and Regulation of ARHGDIB/RhoGDI2 Expression Including the Hypothetical Role of ARHGDIB/RhoGDI2 Autoantibodies in Kidney Transplantation. Transplant Direct 2020; 6:e548. [PMID: 32548242 PMCID: PMC7213606 DOI: 10.1097/txd.0000000000000993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022] Open
Abstract
Challenging and still unsolved problems in kidney transplantation are risk stratification and the treatment of humoral rejection. Antibody-mediated rejection is an important cause of early and chronic rejection. The impact of donor-specific HLA antibodies on antibody-mediated rejection–causing graft damage is well known, but the clinical relevance of non-HLA antibodies remains unclear. Recently, in 2 independent studies, a new correlation was found between the presence of non-HLA anti-Rho guanosine diphosphate dissociation inhibitor 2 (ARHGDIB) antibodies and increased graft failure. RhoGDI2, another name for ARHGDIB, is a negative regulator of the Rho guanosine triphosphate (RhoGTP)ases RhoA, Rac1m, and Cdc42, whose main function is regulating the actin network in a variety of cells. RhoGDI2 is mainly expressed intracellularly, and some expression is observed on the cell surface. Currently, there is no mechanism known to explain this correlation. Additionally, the reason why the antibodies are produced is unknown. In this review, we will address these questions, provide an overview of other diseases in which these antibodies are prevalent, and describe the physiological role of RhoGDI2 itself. If the mechanism and impact of RhoGDI2 antibodies in kidney graft failure are known, improved risk stratification can be provided to decrease the rate of donor kidney graft failure.
Collapse
|
27
|
Urinary MicroRNA-21-5p as Potential Biomarker of Interstitial Fibrosis and Tubular Atrophy (IFTA) in Kidney Transplant Recipients. Diagnostics (Basel) 2020; 10:diagnostics10020113. [PMID: 32092939 PMCID: PMC7168003 DOI: 10.3390/diagnostics10020113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/11/2020] [Accepted: 02/17/2020] [Indexed: 12/29/2022] Open
Abstract
Chronic renal allograft dysfunction (CAD) is a major limiting factor of long-term graft survival. The hallmarks of progressive CAD are interstitial fibrosis and tubular atrophy (IFTA). MicroRNAs are small, regulatory RNAs involved in many immunological processes. In particular, microRNA-21-5p (miR-21) is considered to be strongly associated with pathogenesis regarding tubulointerstitium. The aim of this study was to assess urinary miR-21 expression levels in the kidney transplant recipients and determine their application in the evaluation of IFTA and kidney allograft function. The expression levels of miR-21 were quantified in the urine of 31 kidney transplant recipients with biopsy-assessed IFTA (IFTA 0 + I: n = 17; IFTA II + III: n = 14) by real-time quantitative PCR. Urine samples were collected at the time of protocolar biopsies performed 1 or 2 years after kidney transplantation. MicroRNA-191-5p was used as reference gene. MiR-21 was significantly up-regulated in IFTA II + III group compared to IFTA 0 + I group (p = 0.003). MiR-21 correlated significantly with serum concentration of creatinine (r = 0.52, p = 0.003) and eGFR (r = -0.45; p = 0.01). ROC analysis determined the diagnostic value of miR-21 with an area under curve (AUC) of 0.80 (p = 0.0002), sensitivity of 0.86 and specificity of 0.71. miR-21 is associated with renal allograft dysfunction and IFTA. Therefore, it could be considered as a potential diagnostic, non-invasive biomarker for monitoring renal graft function.
Collapse
|
28
|
Torquetenovirus Serum Load and Long-Term Outcomes in Renal Transplant Recipients. J Clin Med 2020; 9:jcm9020440. [PMID: 32041187 PMCID: PMC7073853 DOI: 10.3390/jcm9020440] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/23/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022] Open
Abstract
Following transplantation, patients must take immunosuppressive medication for life. Torquetenovirus (TTV) is thought to be marker for immunosuppression, and TTV–DNA levels after organ transplantation have been investigated, showing high TTV levels, associated with increased risk of infections, and low TTV levels associated with increased risk of rejection. However, this has been investigated in studies with relatively short follow-up periods. We hypothesized that TTV levels can be used to assess long term outcomes after renal transplantation. Serum samples of 666 renal transplant recipients were tested for TTV DNA. Samples were taken at least one year after renal transplantation, when TTV levels are thought to be relatively stable. Patient data was reviewed for graft failure, all-cause mortality and death due to infectious causes. Our data indicates that high TTV levels, sampled more than one year post-transplantation, are associated with all-cause mortality with a hazard ratio (HR) of 1.12 (95% CI, 1.02–1.23) per log10 increase in TTV viral load, (p = 0.02). Additionally, high TTV levels were also associated with death due to infectious causes (HR 1.20 (95% CI 1.01–1.43), p = 0.04). TTV levels decrease in the years following renal transplantation, but remain elevated longer than previously thought. This study shows that TTV level may aid in predicting long-term outcomes, all-cause mortality and death due to an infectious cause in renal transplant patients sampled over one year post-transplantation.
Collapse
|
29
|
The Causes of Kidney Allograft Failure: More Than Alloimmunity. A Viewpoint Article. Transplantation 2020; 104:e46-e56. [DOI: 10.1097/tp.0000000000003012] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Casiraghi F, Perico N, Gotti E, Todeschini M, Mister M, Cortinovis M, Portalupi V, Plati AR, Gaspari F, Villa A, Introna M, Longhi E, Remuzzi G. Kidney transplant tolerance associated with remote autologous mesenchymal stromal cell administration. Stem Cells Transl Med 2019; 9:427-432. [PMID: 31872574 PMCID: PMC7103624 DOI: 10.1002/sctm.19-0185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Here we report the case of successful immune tolerance induction in a living‐donor kidney transplant recipient remotely treated with autologous bone marrow‐derived mesenchymal stromal cells (MSC). This case report, which to the best of our knowledge is the first in the world in this setting, provides evidence that the modulation of the host immune system with MSC can enable the safe withdrawal of maintenance immunosuppressive drugs while preserving optimal long‐term kidney allograft function.
Collapse
Affiliation(s)
| | - Norberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Eliana Gotti
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Marta Todeschini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marilena Mister
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Monica Cortinovis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Valentina Portalupi
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Anna Rita Plati
- Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Flavio Gaspari
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Alessandro Villa
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Martino Introna
- G. Lanzani Laboratory of Cell Therapy, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Elena Longhi
- Laboratory of Transplant Immunology, UOC Coordinamento Trapianti IRCCS Fondazione Ca' Granda - Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
31
|
Parajuli S, Aziz F, Garg N, Panzer SE, Joachim E, Muth B, Mohamed M, Blazel J, Zhong W, Astor BC, Mandelbrot DA, Djamali A. Histopathological characteristics and causes of kidney graft failure in the current era of immunosuppression. World J Transplant 2019; 9:123-133. [PMID: 31750089 PMCID: PMC6851501 DOI: 10.5500/wjt.v9.i6.123] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The histopathological findings on the failing kidney allograft in the modern era is not well studied. In this study, we present our experience working with kidney transplant recipients with graft failure within one year of the biopsy. AIM To report the histopathological characteristics of failed kidney allografts in the current era of immunosuppression based on the time after transplant, cause of the end-stage renal disease and induction immunosuppressive medications. METHODS In a single-center observational study, we characterized the histopathological findings of allograft biopsies in kidney transplant recipients with graft failure within one year after the biopsy. RESULTS We identified 329 patients with graft failure that met the selection criteria between January 1, 2006 and December 31, 2016. The three most common biopsy findings were interstitial fibrosis and tubular atrophy (IFTA, 53%), acute rejection (AR, 43%) and transplant glomerulopathy (TG, 33%). Similarly, the three most common causes of graft failure based on the primary diagnosis were AR (40%), TG (17%), and IFTA (13%). Most grafts failed within two years of post-transplant (36%). Subsequently, approximately 10%-15% of grafts failed every two years: > 2-4 years (16%), > 4-6 years (13%), > 6-8 years (11%), > 8-10 years (9%) and > 10 years (16%). AR was the most common cause of graft failure in the first six years (48%), whereas TG was the most prevalent cause of graft failure after 6 years (32%) of transplant. CONCLUSION In the current era of immunosuppression, AR is still the most common cause of early graft failure, while TG is the most prevalent cause of late graft failure.
Collapse
Affiliation(s)
- Sandesh Parajuli
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Fahad Aziz
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Neetika Garg
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Sarah E Panzer
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Emily Joachim
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Brenda Muth
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Maha Mohamed
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Justin Blazel
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Weixiong Zhong
- Department of Pathology, University of Wisconsin, Madison, WI 53705, United States
| | - Brad C Astor
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
- Department of Population Health Sciences, University of Wisconsin, Madison, WI 53705, United States
| | - Didier A Mandelbrot
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
| | - Arjang Djamali
- Division of Nephrology, Department of Medicine, University of Wisconsin, Madison, WI 53705, United States
- Division of Transplantation, Department of Surgery, University of Wisconsin, Madison, WI 53705, United States
| |
Collapse
|
32
|
Nair P, Gheith O, Al-Otaibi T, Mostafa M, Rida S, Sobhy I, Halim MA, Mahmoud T, Abdul-Hameed M, Maher A, Emam M. Management of Chronic Active Antibody-Mediated Rejection in Renal Transplant Recipients: Single-Center Experience. EXP CLIN TRANSPLANT 2019; 17:113-119. [PMID: 30777534 DOI: 10.6002/ect.mesot2018.o58] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Data on the management of chronic antibody-mediated rejection after kidney transplantation are limited. We aimed to assess the impact of treatment of biopsy-proven chronic active antibodymediated rejection with combined plasma exchange, intravenous immunoglobulin, and rituximab treatment versus intravenous immunoglobulin alone or conservative management on the evolution of renal function in renal transplant recipients. MATERIALS AND METHODS In this retrospective study, we compared patients diagnosed with chronic active antibody-mediated rejection who were treated with standard of care steroids, intravenous immunoglobulin, plasma exchange, and rituximab (n = 40) at our center versus those who received intravenous immunoglobulin only or just intensified maintenance immunosuppression (n = 28). All patients were followed for 12 months clinically and by laboratory tests for graft and patient outcomes. RESULTS The two groups were matched regarding mean recipient age (41.9 ± 15.4 vs 37.8 ± 15.5 y in patients with conservative versus combined treatment), recipient sex, mean body weight, and the cause of end-stage kidney disease. Most patients and their donors were males. Glomerulonephritis represented the most common cause of end-stage kidney disease in both groups followed by diabetic nephropathy. The type of induction and pretransplant comorbidities were not different between groups (P > .05) except for the significantly higher number of chronic hepatitis C infections in patients who received conservative treatment (P = .007). Mean serum creatinine values before and after treatment of chronic active antibodymediated rejection were comparable between groups (P > .05). Active treatment with heavier immunosuppression (rituximab and plasma exchange) was associated with posttreatment viral (cytomegalovirus and BK virus) and bacterial infections that necessitated more hospitalization (P > .05). However, graft and patient outcomes were significantly better in the active treatment group than in patients with conservative treatment (P = .002 and .028, respectively). CONCLUSIONS Combined treatment of chronic active antibody-mediated rejection with plasma exchange, intravenous immunoglobulin, and rituximab can significantly improve outcomes after renal transplant.
Collapse
Affiliation(s)
- Prasad Nair
- From the Kuwait Ministry of Health, Hamed Al-Essa Organ Transplant Center, Sabah area, Kuwait
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
De novo donor-specific antibody (DSA) formation is a major problem in transplantation, and associated with long-term graft decline and loss as well as sensitization, limiting future transplant options. Forming high-affinity, long-lived antibody responses involves a process called the germinal center (GC) reaction, and requires interaction between several cell types, including GC B cells, T follicular helper (Tfh) and T follicular regulatory (Tfr) cells. T follicular regulatory cells are an essential component of the GC reaction, limiting its size and reducing nonspecific or self-reactive responses.An imbalance between helper function and regulatory function can lead to excessive antibody production. High proportions of Tfh cells have been associated with DSA formation in transplantation; therefore, Tfr cells are likely to play an important role in limiting DSA production. Understanding the signals that govern Tfr cell development and the balance between helper and regulatory function within the GC is key to understanding how these cells might be manipulated to reduce the risk of DSA development.This review discusses the development and function of Tfr cells and their relevance to transplantation. In particular how current and future immunosuppressive strategies might allow us to skew the ratio between Tfr and Tfh cells to increase or decrease the risk of de novo DSA formation.
Collapse
|
34
|
Wunderink HF, De Brouwer CS, Gard L, De Fijter JW, Kroes ACM, Rotmans JI, Feltkamp MCW. Source and Relevance of the BK Polyomavirus Genotype for Infection After Kidney Transplantation. Open Forum Infect Dis 2019; 6:ofz078. [PMID: 30949528 PMCID: PMC6440680 DOI: 10.1093/ofid/ofz078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 02/14/2019] [Indexed: 12/23/2022] Open
Abstract
Background BK polyomavirus (BKPyV)–associated nephropathy (BKPyVAN) is a major threat for kidney transplant recipients (KTRs). The role of specific BKPyV genotypes/serotypes in development of BKPyVAN is poorly understood. Pretransplantation serotyping of kidney donors and recipients and posttransplantation genotyping of viremic recipients, could reveal the clinical relevance of specific BKPyV variants. Methods A retrospective cohort of 386 living kidney donor-recipient pairs was serotyped before transplantation against BKPyV genotype I–IV viral capsid protein 1 antigen, using a novel BKPyV serotyping assay. Replicating BKPyV isolates in viremic KTRs after transplantation were genotyped using real-time polymerase chain reaction and confirmed by means of sequencing. BKPyV serotype and genotype data were used to determine the source of infection and analyze the risk of viremia and BKPyVAN. Results Donor and recipient BKPyV genotype and serotype distribution was dominated by genotype I (>80%), especially Ib, over II, III and IV. Donor serotype was significantly correlated with the replicating genotype in viremic KTRs (P < .001). Individual donor and recipient serotype, serotype (mis)matching and the recipient replicating BKPyV genotype were not associated with development of viremia or BKPyVAN after transplantation. Conclusions BKPyV donor and recipient serotyping and genotyping indicates the donor origin of replicating BKPyV in viremic KTRs but provides no evidence for BKPyV genotype–specific virulence.
Collapse
Affiliation(s)
- H F Wunderink
- Department of Medical Microbiology, Leiden University Medical Center, the Netherlands
| | - C S De Brouwer
- Department of Medical Microbiology, Leiden University Medical Center, the Netherlands
| | - L Gard
- Department of Medical Microbiology, University Medical Center Groningen, the Netherlands
| | - J W De Fijter
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands
| | - A C M Kroes
- Department of Medical Microbiology, Leiden University Medical Center, the Netherlands
| | - J I Rotmans
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands
| | - M C W Feltkamp
- Department of Medical Microbiology, Leiden University Medical Center, the Netherlands
| |
Collapse
|
35
|
Cernoch M, Hruba P, Kollar M, Mrazova P, Stranavova L, Lodererova A, Honsova E, Viklicky O. Intrarenal Complement System Transcripts in Chronic Antibody-Mediated Rejection and Recurrent IgA Nephropathy in Kidney Transplantation. Front Immunol 2018; 9:2310. [PMID: 30356754 PMCID: PMC6189372 DOI: 10.3389/fimmu.2018.02310] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/17/2018] [Indexed: 01/26/2023] Open
Abstract
Background: The complement system activation and regulation have been linked to post-transplant pathologies including chronic antibody mediated rejection (cAMR) and the recurrence of IgA nephropathy (ReIgAN) but distinct mechanisms remain to be elucidated. Methods: In this retrospective single center study, the outcome of kidney transplantation was studied in 150 patients with late histological diagnosis to be either cAMR or ReIgAN, 14 stable kidney grafts at 3 months and finally 11 patients with native kidney IgAN nephropathy. To study a role of complement cascade and regulation in cAMR and ReIgAN, the RNA was extracted from available frozen kidney biopsy samples and using RT-qPCR transcripts of 11 target genes along with clinical data were determined and compared with stable grafts at 3 months protocol biopsies or IgAN native kidney nephropathy. Immunohistologically, CD46 (MCP), and C5 proteins were stained in biopsies. Results: Interestingly, there were no differences in kidney graft survival between cAMR and ReIgAN since transplantation. cAMR was associated with significantly higher intragraft transcripts of C3, CD59, and C1-INH as compared to ReIgAN (p < 0.05). When compared to normal stable grafts, cAMR grafts exhibited higher C3, CD55, CD59, CFH, CFI, and C1-INH (p < 0.01). Moreover, ReIgAN was associated with the increase of CD46, CD55, CD59 (p < 0.01), and CFI (p < 0.05) transcripts compared with native kidney IgAN. Rapid progression of cAMR (failure at 2 years after biopsy) was observed in patients with lower intrarenal CD55 expression (AUC 0.77, 78.6% sensitivity, and 72.7 specificity). There was highly significant association of several complement intrarenal transcripts and the degree of CKD regardless the diagnosis; C3, CD55, CFH, CFI, and C1-INH expressions positively correlated with eGFR (for all p < 0.001). Neither the low mRNA transcripts nor the high mRNA transcripts biopsies were associated with distinct trend in MCP or C5 proteins staining. Conclusions: The intrarenal complement system transcripts are upregulated in progressively deteriorated kidney allografts.
Collapse
Affiliation(s)
- Marek Cernoch
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Petra Hruba
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Marek Kollar
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Petra Mrazova
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Lucia Stranavova
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Alena Lodererova
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Eva Honsova
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Ondrej Viklicky
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia.,Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| |
Collapse
|
36
|
Review: Management of patients with kidney allograft failure. Transplant Rev (Orlando) 2018; 32:178-186. [DOI: 10.1016/j.trre.2018.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/18/2018] [Accepted: 03/21/2018] [Indexed: 12/25/2022]
|
37
|
Moreso F, Crespo M, Ruiz JC, Torres A, Gutierrez-Dalmau A, Osuna A, Perelló M, Pascual J, Torres IB, Redondo-Pachón D, Rodrigo E, Lopez-Hoyos M, Seron D. Treatment of chronic antibody mediated rejection with intravenous immunoglobulins and rituximab: A multicenter, prospective, randomized, double-blind clinical trial. Am J Transplant 2018; 18:927-935. [PMID: 28949089 DOI: 10.1111/ajt.14520] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 02/06/2023]
Abstract
There are no approved treatments for chronic antibody mediated rejection (ABMR). We conducted a multicenter, prospective, randomized, placebo-controlled, double-blind clinical trial to evaluate efficacy and safety of intravenous immunoglobulins (IVIG) combined with rituximab (RTX) (EudraCT 2010-023746-67). Patients with transplant glomerulopathy and anti-HLA donor-specific antibodies (DSA) were eligible. Patients with estimated glomerular filtration rate (eGFR) <20 mL/min per 1.73m2 and/or severe interstitial fibrosis/tubular atrophy were excluded. Patients were randomized to receive IVIG (4 doses of 0.5 g/kg) and RTX (375 mg/m2 ) or a wrapped isovolumetric saline infusion. Primary efficacy variable was the decline of eGFR at one year. Secondary efficacy variables included evolution of proteinuria, renal lesions, and DSA at 1 year. The planned sample size was 25 patients per group. During 2012-2015, 25 patients were randomized (13 to the treatment and 12 to the placebo group). The planned patient enrollment was not achieved because of budgetary constraints and slow patient recruitment. There were no differences between the treatment and placebo groups in eGFR decline (-4.2 ± 14.4 vs. -6.6 ± 12.0 mL/min per 1.73 m2 , P-value = .475), increase of proteinuria (+0.9 ± 2.1 vs. +0.9 ± 2.1 g/day, P-value = .378), Banff scores at one year and MFI of the immunodominant DSA. Safety was similar between groups. These data suggest that the combination of IVIG and RTX is not useful in patients displaying transplant glomerulopathy and DSA.
Collapse
Affiliation(s)
- Francesc Moreso
- Nephrology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Crespo
- Nephrology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Juan C Ruiz
- Nephrology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Armando Torres
- Nephrology Department, Hospital Universitario de Canarias, La Laguna, Spain
| | | | - Antonio Osuna
- Nephrology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Manel Perelló
- Nephrology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Julio Pascual
- Nephrology Department, Hospital del Mar, Parc de Salut Mar, Barcelona, Spain
| | - Irina B Torres
- Nephrology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Emilio Rodrigo
- Nephrology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Marcos Lopez-Hoyos
- Immunology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Daniel Seron
- Nephrology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Nankivell BJ, Shingde M, Keung KL, Fung CLS, Borrows RJ, O'Connell PJ, Chapman JR. The causes, significance and consequences of inflammatory fibrosis in kidney transplantation: The Banff i-IFTA lesion. Am J Transplant 2018; 18:364-376. [PMID: 29194971 DOI: 10.1111/ajt.14609] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/25/2017] [Accepted: 10/28/2017] [Indexed: 01/25/2023]
Abstract
Inflammation within areas of interstitial fibrosis and tubular atrophy (i-IFTA) is associated with adverse outcomes in kidney transplantation. We evaluated i-IFTA in 429 indication- and 2052 protocol-driven biopsy samples from a longitudinal cohort of 362 kidney-pancreas recipients to determine its prevalence, time course, and relationships with T cell-mediated rejection (TCMR), immunosuppression, and outcome. Sequential histology demonstrated that i-IFTA was preceded by cellular interstitial inflammation and followed by IF/TA. The prevalence and intensity of i-IFTA increased with developing chronic fibrosis and correlated with inflammation, tubulitis, and immunosuppression era (P < .001). Tacrolimus era-based immunosuppression was associated with reduced histologic inflammation in unscarred and scarred i-IFTA compartments, ameliorated progression of IF, and increased conversion to inactive IF/TA (compared with cyclosporine era, P < .001). Prior acute (including borderline) TCMR and subclinical TCMR were followed by greater 1-year i-IFTA, remaining predictive by multivariate analysis and independent of humoral markers. One-year i-IFTA was associated with accelerated IF/TA, arterial fibrointimal hyperplasia, and chronic glomerulopathy and with reduced renal function (P < .001 versus no i-IFTA). In summary, i-IFTA is the histologic consequence of active T cell-mediated alloimmunity, representing the interface between inflammation and tubular injury with fibrotic healing. Uncontrolled i-IFTA is associated with adverse structural and functional outcomes.
Collapse
Affiliation(s)
| | - Meena Shingde
- Tissue Pathology and Diagnostic Oncology, Westmead Hospital, Sydney, Australia
| | - Karen L Keung
- Department of Renal Medicine, Westmead Hospital, Sydney, Australia
| | - Caroline L-S Fung
- Tissue Pathology and Diagnostic Oncology, Westmead Hospital, Sydney, Australia
| | | | | | - Jeremy R Chapman
- Department of Renal Medicine, Westmead Hospital, Sydney, Australia
| |
Collapse
|
39
|
Scadden JRW, Sharif A, Skordilis K, Borrows R. Polyoma virus nephropathy in kidney transplantation. World J Transplant 2017; 7:329-338. [PMID: 29312862 PMCID: PMC5743870 DOI: 10.5500/wjt.v7.i6.329] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 11/17/2017] [Accepted: 12/01/2017] [Indexed: 02/05/2023] Open
Abstract
BK virus (BKV) is a polyomavirus that is able to cause renal dysfunction in transplanted grafts via BK virus-associated nephritis (BKVAN). This condition was mis-diagnosed in the past due to clinical and histopthological similarities with acute rejection. Due to the prevalence of the virus in the population, it is an important pathogen in this context, and so it is important to understand how this virus functions and its' relationship with the pathogenesis of BKVN. Screening for BKV often reveals viruria and/or viremia, which then manifests as BKVN, which can be asymptomatic or result in clinical features namely renal dysfunction. The pathogenesis of BKV infection is still unclear and needs to be further investigated; nevertheless there are a variety of hypotheses that indicate that there are a host of factors that play important roles. Treatments for BKVAN include a reduction in immunosuppression, the use of antiviral therapy or the combination of both treatment options.
Collapse
Affiliation(s)
- Jacob RW Scadden
- University of Birmingham, Edgbaston, Birmingham B15 2TH, United Kingdom
| | - Adnan Sharif
- Department of Kidney Transplantation, Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, United Kingdom
| | - Kassi Skordilis
- Department of Renal Histopathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, United Kingdom
| | - Richard Borrows
- Department of Kidney Transplantation, Queen Elizabeth Hospital Birmingham, Birmingham B15 2TH, United Kingdom
| |
Collapse
|
40
|
de Leur K, Dor FJMF, Dieterich M, van der Laan LJW, Hendriks RW, Baan CC. IL-21 Receptor Antagonist Inhibits Differentiation of B Cells toward Plasmablasts upon Alloantigen Stimulation. Front Immunol 2017; 8:306. [PMID: 28373876 PMCID: PMC5357809 DOI: 10.3389/fimmu.2017.00306] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/03/2017] [Indexed: 12/04/2022] Open
Abstract
Interaction between T follicular helper (Tfh) cells and B cells is complex and involves various pathways, including the production of IL-21 by the Tfh cells. Secretion of IL-21 results in B cell differentiation toward immunoglobulin-producing plasmablasts. In patients after kidney transplantation, the formation of alloantibodies produced by donor antigen-activated B cells are a major cause of organ failure. In this allogeneic response, the role of IL-21-producing Tfh cells that regulate B cell differentiation is unknown. Here, we tested, in an alloantigen-driven setting, whether Tfh cell help signals control B cell differentiation with its dependency on IL-21. Pre-transplantation patient PBMCs were sorted into pure CD4posCXCR5pos Tfh cells and CD19posCD27pos memory B cells and stimulated with donor antigen in the presence or absence of an IL-21 receptor (IL-21R) antagonist (αIL-21R). Donor antigen stimulation initiated expression of the activation markers inducible co-stimulator (ICOS) and programmed death 1 (PD-1) on Tfh cells and a shift toward a mixed Tfh2 and Tfh17 phenotype. The memory B cells underwent class switch recombination and differentiated toward IgM- and IgG-producing plasmablasts. In the presence of αIL-21R, a dose-dependent inhibition of STAT3 phosphorylation was measured in both T and B cells. Blockade of the IL-21R did not have an effect on PD-1 and ICOS expression on Tfh cells but significantly inhibited B cell differentiation. The proportion of plasmablasts decreased by 78% in the presence of αIL-21R. Moreover, secreted IgM and IgG2 levels were significantly lower in the presence of αIL-21R. In conclusion, our results demonstrate that IL-21 produced by alloantigen-activated Tfh cells controls B cell differentiation toward antibody producing plasmablasts. The IL-21R might, therefore, be a useful target in organ transplantation to prevent antigen-driven immune responses leading to graft failure.
Collapse
Affiliation(s)
- Kitty de Leur
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands; Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Frank J M F Dor
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center , Rotterdam , Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Erasmus MC, University Medical Center , Rotterdam , Netherlands
| | - Luc J W van der Laan
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center , Rotterdam , Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center , Rotterdam , Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Erasmus MC, University Medical Center , Rotterdam , Netherlands
| |
Collapse
|