151
|
Goutaudier V, Perrochia H, Mucha S, Bonnet M, Delmas S, Garo F, Garrigue V, Lepreux S, Pernin V, Serre JE, Szwarc I, Merville P, Ramounau-Pigot A, René C, Visentin J, Morgan BP, Frémeaux-Bacchi V, Mourad G, Couzi L, Le Quintrec M. C5b9 Deposition in Glomerular Capillaries Is Associated With Poor Kidney Allograft Survival in Antibody-Mediated Rejection. Front Immunol 2019; 10:235. [PMID: 30906289 PMCID: PMC6418012 DOI: 10.3389/fimmu.2019.00235] [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: 11/16/2018] [Accepted: 01/28/2019] [Indexed: 12/17/2022] Open
Abstract
C4d deposition in peritubular capillaries (PTC) reflects complement activation in antibody-mediated rejection (ABMR) of kidney allograft. However, its association with allograft survival is controversial. We hypothesized that capillary deposition of C5b9-indicative of complement-mediated injury-is a severity marker of ABMR. This pilot study aimed to determine the frequency, location and prognostic impact of these deposits in ABMR. We retrospectively selected patients diagnosed with ABMR in two French transplantation centers from January 2005 to December 2014 and performed C4d and C5b9 staining by immunohistochemistry. Fifty-four patients were included. Median follow-up was 52.5 (34.25-73.5) months. Thirteen patients (24%) had C5b9 deposits along glomerular capillaries (GC). Among these, seven (54%) had a global and diffuse staining pattern. Twelve of the C5b9+ patients also had deposition of C4d in GC and PTC. C4d deposits along GC and PTC were not associated with death-censored allograft survival (p = 0.42 and 0.69, respectively). However, death-censored allograft survival was significantly lower in patients with global and diffuse deposition of C5b9 in GC than those with a segmental pattern or no deposition (median survival after ABMR diagnosis, 6 months, 40.5 months and 44 months, respectively; p = 0.015). Double contour of glomerular basement membrane was diagnosed earlier after transplantation in C5b9+ ABMR than in C5b9- ABMR (median time after transplantation, 28 vs. 85 months; p = 0.058). In conclusion, we identified a new pattern of C5b9+ ABMR, associated with early onset of glomerular basement membrane duplication and poor allograft survival. Complement inhibitors might be a therapeutic option for this subgroup of patients.
Collapse
Affiliation(s)
- Valentin Goutaudier
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Hélène Perrochia
- Department of Pathology, Gui de Chauliac Hospital, Montpellier University Hospital, Montpellier, France
| | - Simon Mucha
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Pellegrin Hospital, Bordeaux University Hospital, Bordeaux, France
| | - Marie Bonnet
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Sylvie Delmas
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Florian Garo
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Valérie Garrigue
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Sébastien Lepreux
- Department of Pathology, Pellegrin Hospital, Bordeaux University Hospital, Bordeaux, France
| | - Vincent Pernin
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France.,INSERM U1183, Institute for Regenerative Medicine and Biotherapy, Saint-Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Jean-Emmanuel Serre
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Ilan Szwarc
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Pierre Merville
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Pellegrin Hospital, Bordeaux University Hospital, Bordeaux, France.,UMR CNRS 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Annie Ramounau-Pigot
- Department of Immunology, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Céline René
- Department of Immunology, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France
| | - Jonathan Visentin
- UMR CNRS 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Department of Immunology and Immunogenetics, Pellegrin Hospital, Bordeaux University Hospital, Bordeaux, France
| | - Bryan Paul Morgan
- School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | | | - Georges Mourad
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Pellegrin Hospital, Bordeaux University Hospital, Bordeaux, France.,UMR CNRS 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Moglie Le Quintrec
- University of Montpellier, Department of Nephrology, Dialysis and Transplantation, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France.,INSERM U1183, Institute for Regenerative Medicine and Biotherapy, Saint-Eloi Hospital, Montpellier University Hospital, Montpellier, France
| |
Collapse
|
152
|
Senev A, Coemans M, Lerut E, Van Sandt V, Daniëls L, Kuypers D, Sprangers B, Emonds MP, Naesens M. Histological picture of antibody-mediated rejection without donor-specific anti-HLA antibodies: Clinical presentation and implications for outcome. Am J Transplant 2019; 19:763-780. [PMID: 30107078 DOI: 10.1111/ajt.15074] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/31/2018] [Accepted: 08/07/2018] [Indexed: 01/25/2023]
Abstract
In this cohort study (n = 935 transplantations), we investigated the phenotype and risk of graft failure in patients with histological criteria for antibody-mediated rejection (ABMR) in the absence of circulating donor-specific anti-human leukocyte antigen (HLA) antibodies (DSA), and compared this to patients with definite ABMR and HLA-DSA-positivity. The histological picture did not differ between HLA-DSA-positive (n = 85) and HLA-DSA-negative (n = 123) cases of ABMR histology, apart from increased complement split product 4d (C4d) deposition in the peritubular capillaries in HLA-DSA-positive cases. Histology of ABMR without HLA-DSA was more transient than DSA-positive ABMR, and patients with ABMR histology without HLA-DSA had graft survival superior to that of HLA-DSA-positive patients, independent of concomitant T cell-mediated rejection (38.2%) or borderline changes (17.9%). Multivariate analysis showed that the risk of graft failure was not higher in patients with histological picture of ABMR (ABMRh ) in the absence of HLA-DSA, compared to patients without ABMRh . Despite an association between C4d deposition and HLA-DSA-positivity, using C4d deposition as alternative for the DSA criterion in the diagnosis of ABMR, as proposed in Banff 2017, did not contribute to the prognosis of graft function and graft failure. We concluded that biopsies with ABMRh but without detectable HLA-DSA represent a distinct, often transient phenotype with superior allograft survival.
Collapse
Affiliation(s)
- Aleksandar Senev
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium.,Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Maarten Coemans
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium
| | - Evelyne Lerut
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Vicky Van Sandt
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Liesbeth Daniëls
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Dirk Kuypers
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Marie-Paule Emonds
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium.,Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
153
|
Timofeeva OA. Donor-Specific HLA Antibodies as Biomarkers of Transplant Rejection. Clin Lab Med 2019; 39:45-60. [DOI: 10.1016/j.cll.2018.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
154
|
Hanaoka K, Maeda M, Tsujimoto S, Oshima S, Fukahori H, Nakamura K, Noto T, Higashi Y, Hirose J, Takakura S, Morokata T. Benefits of a loading dose of tacrolimus on graft survival of kidney transplants in nonhuman primates. Transpl Immunol 2019. [DOI: 10.1016/j.trim.2018.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
155
|
Marsh CL, Kurian SM, Rice JC, Whisenant TC, David J, Rose S, Schieve C, Lee D, Case J, Barrick B, Peddi VR, Mannon RB, Knight R, Maluf D, Mandelbrot D, Patel A, Friedewald JJ, Abecassis MM, First MR. Application of TruGraf v1: A Novel Molecular Biomarker for Managing Kidney Transplant Recipients With Stable Renal Function. Transplant Proc 2019; 51:722-728. [PMID: 30979456 DOI: 10.1016/j.transproceed.2019.01.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/10/2018] [Accepted: 01/17/2019] [Indexed: 01/22/2023]
Abstract
TruGraf v1 is a laboratory-developed DNA microarray-based gene expression blood test to enable proactive noninvasive serial assessment of kidney transplant recipients with stable renal function. It has been previously validated in patients identified as Transplant eXcellence (TX: stable serum creatinine, normal biopsy results, indicative of immune quiescence), and not-TX (renal dysfunction and/or rejection on biopsy results). TruGraf v1 is intended for use in subjects with stable renal function to measure the immune status as an alternative to invasive, expensive, and risky surveillance biopsies. MATERIALS AND METHODS In this study, simultaneous blood tests and clinical assessments were performed in 192 patients from 7 transplant centers to evaluate TruGraf v1. The molecular testing laboratory was blinded to renal function and biopsy results. RESULTS Overall, TruGraf v1 accuracy (concordance between TruGraf v1 result and clinical and/or histologic assessment) was 74% (142/192), and a result of TX was accurate in 116 of 125 (93%). The negative predictive value for TruGraf v1 was 90%, with a sensitivity 74% and specificity of 73%. Results did not significantly differ in patients with a biopsy-confirmed diagnosis vs those without a biopsy. CONCLUSIONS TruGraf v1 can potentially support a clinical decision enabling unnecessary surveillance biopsies with high confidence, making it an invaluable addition to the transplant physician's tool kit for managing patients. TruGraf v1 testing can potentially avoid painful and risky invasive biopsies, reduce health care costs, and enable frequent assessment of patients with stable renal function to confirm the presence of immune quiescence in the peripheral blood.
Collapse
Affiliation(s)
- C L Marsh
- Scripps Center for Organ Transplantation, La Jolla, California, United States; Scripps Clinic Bio-Repository and Transplantation Research, La Jolla, California, United States.
| | - S M Kurian
- Scripps Clinic Bio-Repository and Transplantation Research, La Jolla, California, United States
| | - J C Rice
- Scripps Center for Organ Transplantation, La Jolla, California, United States
| | - T C Whisenant
- University of California, San Diego, School of Medicine, Center for Computational Biology and Bioinformatics, La Jolla, California, United States
| | - J David
- Transplant Genomics Inc, Mansfield, Massachusetts, United States
| | - S Rose
- Transplant Genomics Inc, Mansfield, Massachusetts, United States
| | - C Schieve
- Transplant Genomics Inc, Mansfield, Massachusetts, United States
| | - D Lee
- Transplant Genomics Inc, Mansfield, Massachusetts, United States
| | - J Case
- Scripps Clinic Bio-Repository and Transplantation Research, La Jolla, California, United States
| | - B Barrick
- Scripps Clinic Bio-Repository and Transplantation Research, La Jolla, California, United States
| | - V R Peddi
- California Pacific Medical Center, San Francisco, California, United States
| | - R B Mannon
- University of Alabama School of Medicine, Birmingham, Alabama, United States
| | - R Knight
- Houston Methodist Hospital, Houston, Texas, United States
| | - D Maluf
- University of Virginia, Charlottesville, Virginia, United States
| | - D Mandelbrot
- University of Wisconsin, Madison, Wisconsin, United States
| | - A Patel
- Henry Ford Hospital, Detroit, Michigan, United States
| | - J J Friedewald
- Comprehensive Transplant Center, Northwestern University, Chicago, Illionis, United States
| | - M M Abecassis
- Comprehensive Transplant Center, Northwestern University, Chicago, Illionis, United States
| | - M R First
- Transplant Genomics Inc, Mansfield, Massachusetts, United States; Comprehensive Transplant Center, Northwestern University, Chicago, Illionis, United States
| |
Collapse
|
156
|
Naesens M. The special relativity of noninvasive biomarkers for acute rejection. Am J Transplant 2019; 19:5-8. [PMID: 30125470 DOI: 10.1111/ajt.15078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/10/2018] [Accepted: 08/12/2018] [Indexed: 01/25/2023]
Affiliation(s)
- Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
157
|
Friedewald JJ, Kurian SM, Heilman RL, Whisenant TC, Poggio ED, Marsh C, Baliga P, Odim J, Brown MM, Ikle DN, Armstrong BD, charette JI, Brietigam SS, Sustento-Reodica N, Zhao L, Kandpal M, Salomon DR, Abecassis MM. Development and clinical validity of a novel blood-based molecular biomarker for subclinical acute rejection following kidney transplant. Am J Transplant 2019; 19:98-109. [PMID: 29985559 PMCID: PMC6387870 DOI: 10.1111/ajt.15011] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/12/2018] [Accepted: 07/03/2018] [Indexed: 01/25/2023]
Abstract
Noninvasive biomarkers are needed to monitor stable patients after kidney transplant (KT), because subclinical acute rejection (subAR), currently detectable only with surveillance biopsies, can lead to chronic rejection and graft loss. We conducted a multicenter study to develop a blood-based molecular biomarker for subAR using peripheral blood paired with surveillance biopsies and strict clinical phenotyping algorithms for discovery and validation. At a predefined threshold, 72% to 75% of KT recipients achieved a negative biomarker test correlating with the absence of subAR (negative predictive value: 78%-88%), while a positive test was obtained in 25% to 28% correlating with the presence of subAR (positive predictive value: 47%-61%). The clinical phenotype and biomarker independently and statistically correlated with a composite clinical endpoint (renal function, biopsy-proved acute rejection, ≥grade 2 interstitial fibrosis, and tubular atrophy), as well as with de novo donor-specific antibodies. We also found that <50% showed histologic improvement of subAR on follow-up biopsies despite treatment and that the biomarker could predict this outcome. Our data suggest that a blood-based biomarker that reduces the need for the indiscriminate use of invasive surveillance biopsies and that correlates with transplant outcomes could be used to monitor KT recipients with stable renal function, including after treatment for subAR, potentially improving KT outcomes.
Collapse
Affiliation(s)
| | | | | | - Thomas C. Whisenant
- UC San Diego Center for Computational Biology & Bioinformatics, San Diego, CA, USA
| | | | | | | | - Jonah Odim
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Merideth M. Brown
- National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | | | | | - jane I. charette
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | - Lihui Zhao
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Manoj Kandpal
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | |
Collapse
|
158
|
Chancharoenthana W, Leelahavanichkul A, Wattanatorn S, Avihingsanon Y, Praditpornsilpa K, Eiam-Ong S, Townamchai N. Alteration of urinary neutrophil gelatinase-associated lipocalin as a predictor of tacrolimus-induced chronic renal allograft fibrosis in tacrolimus dose adjustments following kidney transplantation. PLoS One 2018; 13:e0209708. [PMID: 30576367 PMCID: PMC6303063 DOI: 10.1371/journal.pone.0209708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/10/2018] [Indexed: 01/05/2023] Open
Abstract
Despite tacrolimus (TAC) drug-level monitoring, TAC-induced chronic renal allograft fibrosis remains an important problem. This study investigated the potential of urinary neutrophil gelatinase–associated lipocalin (uNGAL) as a chronic renal allograft fibrosis biomarker in a two-phase study (proof of concept and cohort). In the proof of concept stage of the study, increased TAC-doses at 3 days after dose adjustment compared with the baseline were associated with elevated uNGAL (+ΔuNGAL) and urinary interleukin 18 (IL-18), but normal serum creatinine (SCr), despite the therapeutic trough levels of TAC. In the cohort study, the patients with elevated uNGAL post-recruitment in comparison with the baseline (+ΔuNGAL) was associated with the more severe renal allograft fibrosis from renal pathology of the protocol biopsy at 12 months post kidney transplantation (post-KT). A cut-off value of uNGAL ≥ 125.2 ng/mL during a 3, 6, 9 and 12 months post-KT was associated with a higher fibrosis score, with an area under the receiver operating characteristics curve of 0.80 (95% confidence interval [CI] 0.72 to 0.88, p < 0.0001) and a hazard ratio (HR) of 2.54 (95% CI 1.45 to 9.33; p < 0.001). We conclude that uNGAL is a sensitive biomarker of TAC induced subtle renal injury and TAC-induced chronic renal allograft fibrosis. We propose that uNGAL measurements, in addition to trough levels of TAC, should be used to predict TAC-induced chronic renal allograft fibrosis in the recipients of KT.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Division of Nephrology, Department of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellent Center of Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- * E-mail:
| | - Asada Leelahavanichkul
- Immunology Unit, Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Salin Wattanatorn
- Division of Nephrology, Department of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellent Center of Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Yingyos Avihingsanon
- Division of Nephrology, Department of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellent Center of Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | | | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natavudh Townamchai
- Division of Nephrology, Department of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellent Center of Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
159
|
The Treatment of Antibody-Mediated Rejection in Kidney Transplantation: An Updated Systematic Review and Meta-Analysis. Transplantation 2018; 102:557-568. [PMID: 29315141 DOI: 10.1097/tp.0000000000002049] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Current treatments for antibody-mediated rejection (AMR) in kidney transplantation are based on low-quality data from a small number of controlled trials. Novel agents targeting B cells, plasma cells, and the complement system have featured in recent studies of AMR. METHODS We conducted a systematic review and meta-analysis of controlled trials in kidney transplant recipients using Medline, EMBASE, and CENTRAL from inception to February 2017. RESULTS Of 14 380 citations, we identified 21 studies, including 10 randomized controlled trials, involving 751 participants. Since the last systematic review conducted in 2011, we found nine additional studies evaluating plasmapheresis + intravenous immunoglobulin (IVIG) (two), rituximab (two), bortezomib (two), C1 inhibitor (two), and eculizumab (one). Risk of bias was serious or unclear overall and evidence quality was low for the majority of treatment strategies. Sufficient RCTs for pooled analysis were available only for antibody removal, and here there was no significant difference between groups for graft survival (HR 0.76; 95% CI 0.35-1.63; P = 0.475). Studies showed important heterogeneity in treatments, definition of AMR, quality, and follow-up. Plasmapheresis and IVIG were used as standard-of-care in recent studies, and to this combination, rituximab seemed to add little or no benefit. Insufficient data are available to assess the efficacy of bortezomib and complement inhibitors. CONCLUSION Newer studies evaluating rituximab showed little or no difference to early graft survival, and the efficacy of bortezomib and complement inhibitors for the treatment of AMR remains unclear. Despite the evidence uncertainty, plasmapheresis and IVIG have become standard-of-care for the treatment of acute AMR.
Collapse
|
160
|
Early Diagnosis and Treatment of Subclinical AMR Is Vital for Improving Clinical Outcomes. Transplantation 2018; 103:1542-1543. [PMID: 30507743 DOI: 10.1097/tp.0000000000002567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
161
|
Advancing Transplantation: New Questions, New Possibilities in Kidney and Liver Transplantation. Transplantation 2018; 101 Suppl 2S:S1-S41. [PMID: 28125449 DOI: 10.1097/tp.0000000000001563] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
162
|
Affiliation(s)
- Alexandre Loupy
- From the Paris Translational Research Center for Organ Transplantation, INSERM, Unité Mixte de Recherche S970 (A.L., C.L.), the Kidney Transplant Department, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) (A.L.), and the Kidney Transplant Department, Saint-Louis Hospital, AP-HP (C.L.) - all in Paris
| | - Carmen Lefaucheur
- From the Paris Translational Research Center for Organ Transplantation, INSERM, Unité Mixte de Recherche S970 (A.L., C.L.), the Kidney Transplant Department, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) (A.L.), and the Kidney Transplant Department, Saint-Louis Hospital, AP-HP (C.L.) - all in Paris
| |
Collapse
|
163
|
Seifert ME, Yanik MV, Feig DI, Hauptfeld-Dolejsek V, Mroczek-Musulman EC, Kelly DR, Rosenblum F, Mannon RB. Subclinical inflammation phenotypes and long-term outcomes after pediatric kidney transplantation. Am J Transplant 2018; 18:2189-2199. [PMID: 29766640 PMCID: PMC6436389 DOI: 10.1111/ajt.14933] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/02/2018] [Accepted: 05/06/2018] [Indexed: 01/25/2023]
Abstract
The implementation of surveillance biopsies in pediatric kidney transplantation remains controversial. Surveillance biopsies detect subclinical injury prior to clinical dysfunction, which could allow for early interventions that prolong allograft survival. We conducted a single-center retrospective cohort study of 120 consecutive pediatric kidney recipients, of whom 103 had surveillance biopsies ≤6 months posttransplant. We tested the hypothesis that subclinical inflammation (borderline or T cell-mediated rejection without clinical dysfunction) is associated with a 5-year composite endpoint of acute rejection and allograft failure. Overall, 36% of subjects had subclinical inflammation, which was associated with increased hazard for the composite endpoint (adjusted hazard ratio 2.89 [1.27, 6.57]; P < .01). Subjects with treated vs untreated subclinical borderline rejection had a lower incidence of the composite endpoint (41% vs 67%; P < .001). Subclinical vascular injury (subclinical inflammation with Banff arteritis score > 0) had a 78% incidence of the composite endpoint vs 11% in subjects with no major surveillance abnormalities (P < .001). In summary, we showed that subclinical inflammation phenotypes were prevalent in pediatric kidney recipients without clinical dysfunction and were associated with increased acute rejection and allograft failure. Once prospectively validated, our data would support implementation of surveillance biopsies as standard of care in pediatric kidney transplantation.
Collapse
Affiliation(s)
- Michael E. Seifert
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama
| | - Megan V. Yanik
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama
| | - Daniel I. Feig
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama
| | - Vera Hauptfeld-Dolejsek
- Department of Surgery, University of Alabama School of Medicine, Birmingham, Alabama,Comprehensive Transplant Institute, University of Alabama School of Medicine, Birmingham, Alabama
| | - Elizabeth C. Mroczek-Musulman
- Children’s of Alabama, Birmingham, Alabama,Department of Pathology, University of Alabama School of Medicine, Birmingham, Alabama
| | - David R. Kelly
- Children’s of Alabama, Birmingham, Alabama,Department of Pathology, University of Alabama School of Medicine, Birmingham, Alabama
| | - Frida Rosenblum
- Department of Pathology, University of Alabama School of Medicine, Birmingham, Alabama
| | - Roslyn B. Mannon
- Department of Surgery, University of Alabama School of Medicine, Birmingham, Alabama,Comprehensive Transplant Institute, University of Alabama School of Medicine, Birmingham, Alabama,Department of Medicine, University of Alabama School of Medicine, Birmingham, Alabama
| |
Collapse
|
164
|
Mincham CM, Gibson IW, Sharma A, Wiebe C, Mandal R, Rush D, Nickerson P, Ho J, Wishart DS, Blydt-Hansen TD. Evolution of renal function and urinary biomarker indicators of inflammation on serial kidney biopsies in pediatric kidney transplant recipients with and without rejection. Pediatr Transplant 2018; 22:e13202. [PMID: 29696778 DOI: 10.1111/petr.13202] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2018] [Indexed: 01/06/2023]
Abstract
Urinary CXCL10 and metabolites are biomarkers independently associated with TCMR. We sought to test whether these biomarkers fluctuate in association with histological severity of TCMR over short time frames. Forty-nine pairs of renal biopsies obtained 1-3 months apart from 40 pediatric renal transplant recipients were each scored for TCMR acuity score (i + t; Banff criteria). Urinary CXCL10:Cr and TCMR MDS were obtained at each biopsy and were tested for association with changes between biopsies in acuity, estimated GFR (ΔeGFR), and 12-month ΔeGFR. Sequential biopsies were obtained 1.8 ± 0.8 months apart. Biopsy 1 was usually obtained under protocol (75%), and 62% percent had evidence of TCMR. Using each biopsy pair for comparison, ΔeGFR did not predict change in acuity. By contrast, change in acuity was significantly correlated with change in urinary CXCL10:Cr (ρ 0.45, P = .003) and MDS (ρ 0.29, P = .04) between biopsies. The 12-month ΔeGFR was not predicted by TCMR acuity or CXCL10:Cr at Biopsy 2; however, an inverse correlation was seen with urinary MDS (ρ -0.35; P = .02). Changes in eGFR correlate poorly with evolving TCMR acuity on histology. Urinary biomarkers may be superior for non-invasive monitoring of rejection, including histological response to therapy, and may be prognostic for medium-term function.
Collapse
Affiliation(s)
- Christine M Mincham
- Department of Pediatrics, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada
| | - Ian W Gibson
- Department of Pathology, University of Manitoba, Health Sciences Center, Winnipeg, MB, Canada
| | - Atul Sharma
- Department of Pediatrics and Child Health, University of Manitoba, Children's Hospital at Health Sciences Center, Winnipeg, MB, Canada
| | - Chris Wiebe
- Department of Internal Medicine, Section of Nephrology, University of Manitoba, Health Sciences Center, Winnipeg, MB, Canada
| | - Rupasri Mandal
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - David Rush
- Department of Internal Medicine, Section of Nephrology, University of Manitoba, Health Sciences Center, Winnipeg, MB, Canada
| | - Peter Nickerson
- Department of Internal Medicine, Section of Nephrology, University of Manitoba, Health Sciences Center, Winnipeg, MB, Canada
| | - Julie Ho
- Department of Internal Medicine, Section of Nephrology, University of Manitoba, Health Sciences Center, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - David S Wishart
- The Metabolomics Innovation Center, University of Alberta, Edmonton, AB, Canada
| | - Tom D Blydt-Hansen
- Department of Pediatrics, University of British Columbia, BC Children's Hospital, Vancouver, BC, Canada
| |
Collapse
|
165
|
Couvrat-Desvergnes G, Foucher Y, Le Borgne F, Dion A, Mourad G, Garrigue V, Legendre C, Rostaing L, Kamar N, Kessler M, Ladrière M, Morelon E, Buron F, Giral M, Dantan E. Comparison of graft and patient survival according to the transplantation centre policy for 1-year screening biopsy among stable kidney recipients: a propensity score-based study. Nephrol Dial Transplant 2018; 34:703-711. [DOI: 10.1093/ndt/gfy221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Grégoire Couvrat-Desvergnes
- Department of Nephrology, Dialysis and Transplantation, Departmental Hospital of Vendée, La Roche-sur-Yon, France
- Centre de Recherche en Transplantation et Immunologie INSERM UMR1064, Université de Nantes, Centre Hospitalier Universitaire de Nantes, RTRS “Centaure”, Nantes, France
| | - Yohann Foucher
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Florent Le Borgne
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
- IDBC/A2com, Pacé, France
| | - Angelina Dion
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
| | - Georges Mourad
- Department of Nephrology, Dialysis and Transplantation, Lapeyronie University Hospital, Montpellier, France
| | - Valérie Garrigue
- Department of Nephrology, Dialysis and Transplantation, Lapeyronie University Hospital, Montpellier, France
| | - Christophe Legendre
- Kidney Transplant Center, Necker University Hospital, APHP, RTRS “Centaure”, Paris Descartes and Sorbonne Paris Cité Universities, Paris, France
| | - Lionel Rostaing
- Department of Nephrology, Dialysis, and Organ Transplantation, Rangueil University Hospital and University Paul Sabatier, Toulouse, France
| | - Nassim Kamar
- Department of Nephrology, Dialysis, and Organ Transplantation, Rangueil University Hospital and University Paul Sabatier, Toulouse, France
| | - Michèle Kessler
- Department of Renal Transplantation, Brabois University Hospital, Nancy, France
| | - Marc Ladrière
- Department of Renal Transplantation, Brabois University Hospital, Nancy, France
| | - Emmanuel Morelon
- Department of Nephrology, Transplantation and Clinic Immunology, RTRS “Centaure”, Edouard Herriot University Hospital, Hospices Civils, Lyon, France
| | - Fanny Buron
- Department of Nephrology, Transplantation and Clinic Immunology, RTRS “Centaure”, Edouard Herriot University Hospital, Hospices Civils, Lyon, France
| | - Magali Giral
- Centre de Recherche en Transplantation et Immunologie INSERM UMR1064, Université de Nantes, Centre Hospitalier Universitaire de Nantes, RTRS “Centaure”, Nantes, France
- Centre d’Investigation Clinique en Biothérapie, Labex Transplantex, Nantes, France
| | - Etienne Dantan
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
| |
Collapse
|
166
|
Mehta R, Bhusal S, Randhawa P, Sood P, Cherukuri A, Wu C, Puttarajappa C, Hoffman W, Shah N, Mangiola M, Zeevi A, Tevar AD, Hariharan S. Short-term adverse effects of early subclinical allograft inflammation in kidney transplant recipients with a rapid steroid withdrawal protocol. Am J Transplant 2018; 18:1710-1717. [PMID: 29247472 DOI: 10.1111/ajt.14627] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/03/2017] [Accepted: 12/10/2017] [Indexed: 01/25/2023]
Abstract
The impact of subclinical inflammation (SCI) noted on early kidney allograft biopsies remains unclear. This study evaluated the outcome of SCI noted on 3-month biopsy. A total of 273/363 (75%) kidney transplant recipients with a functioning kidney underwent allograft biopsies 3-months posttransplant. Among those with stable allograft function at 3 months, 200 biopsies that did not meet the Banff criteria for acute rejection were identified. These were Group I: No Inflammation (NI, n = 71) and Group II: Subclinical Inflammation (SCI, n = 129). We evaluated differences in kidney function at 24-months and allograft histology score at 12-month biopsy. SCI patients had a higher serum creatinine (1.6 ± 0.7 vs 1.38 ± 0.45; P = .02) at 24-months posttransplant, and at last follow-up at a mean of 42.5 months (1.69 ± 0.9 vs 1.46 ± 0.5 mg/dL; P = .027). The allograft chronicity score (ci + ct + cg + cv) at 12-months posttransplant was higher in the SCI group (2.4 ± 1.35 vs 1.9 ± 1.2; P = .02). The incidence of subsequent rejections within the first year in SCI and NI groups was 24% vs 10%, respectively (P = .015). De novo donor-specific antibody within 12 months was more prevalent in the SCI group (12/129 vs 1/71, P = .03). SCI is likely not a benign finding and may have long-term implications for kidney allograft function.
Collapse
Affiliation(s)
- Rajil Mehta
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sushma Bhusal
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Parmjeet Randhawa
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Puneet Sood
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Aravind Cherukuri
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Christine Wu
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Chethan Puttarajappa
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - William Hoffman
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nirav Shah
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Massimo Mangiola
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adriana Zeevi
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Amit D Tevar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sundaram Hariharan
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
167
|
Alkadi MM, Kim J, Aull MJ, Schwartz JE, Lee JR, Watkins A, Lee JB, Dadhania DM, Seshan SV, Serur D, Kapur S, Suthanthiran M, Hartono C, Muthukumar T. Kidney allograft failure in the steroid-free immunosuppression era: A matched case-control study. Clin Transplant 2018; 31. [PMID: 28921709 DOI: 10.1111/ctr.13117] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
We studied the causes and predictors of death-censored kidney allograft failure among 1670 kidney recipients transplanted at our center in the corticosteroid-free maintenance immunosuppression era. As of January 1, 2012, we identified 137 recipients with allograft failure; 130 of them (cases) were matched 1-1 for recipient age, calendar year of transplant, and donor type with 130 recipients with functioning grafts (controls). Median time to allograft failure was 29 months (interquartile range: 18-51). Physician-validated and biopsy-confirmed categories of allograft failure were as follows: acute rejection (21%), glomerular disease (19%), transplant glomerulopathy (13%), interstitial fibrosis tubular atrophy (10%), and polyomavirus-associated nephropathy (7%). Graft failures were attributed to medical conditions in 21% and remained unresolved in 9%. Donor race, donor age, human leukocyte antigen mismatches, serum creatinine, urinary protein, acute cellular rejection, acute antibody-mediated rejection, BK viremia, and CMV viremia were associated with allograft failure. Independent predictors of allograft failure were acute cellular rejection (odds ratio: 18.31, 95% confidence interval: 5.28-63.45) and urine protein ≥1 g/d within the first year post-transplantation (5.85, 2.37-14.45). Serum creatinine ≤1.5 mg/dL within the first year post-transplantation reduced the odds (0.29, 0.13-0.64) of allograft failure. Our study has identified modifiable risk factors to reduce the burden of allograft failure.
Collapse
Affiliation(s)
- Mohamad M Alkadi
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Division of Nephrology, Department of Medicine, Hamad Medical Corporation, Doha, Qatar
| | - Jim Kim
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Meredith J Aull
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Joseph E Schwartz
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - John R Lee
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Anthony Watkins
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Jun B Lee
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,The Rogosin Institute, New York, NY, USA
| | - Darshana M Dadhania
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Surya V Seshan
- Department of Pathology, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - David Serur
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,The Rogosin Institute, New York, NY, USA
| | - Sandip Kapur
- Division of Transplantation Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Manikkam Suthanthiran
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Choli Hartono
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,The Rogosin Institute, New York, NY, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| |
Collapse
|
168
|
Giral M, Renaudin K, Naesens M, Luning R, Anglicheau D, Morelon E, Huneau A, Paul C, Brouard S, Couvrat-Desvergnes G, Foucher Y, Dantan E. The 1-year Renal Biopsy Index: a scoring system to drive biopsy indication at 1-year post-kidney transplantation. Transpl Int 2018; 31:947-955. [PMID: 29893433 DOI: 10.1111/tri.13290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/26/2018] [Accepted: 06/07/2018] [Indexed: 11/27/2022]
Abstract
Surveillance biopsies after renal transplantation remain debatable. To drive the decision of such intervention, we propose a predictive score of abnormal histology at 1-year post-transplantation, named 1-year Renal Biopsy Index (1-RBI). We studied 466 kidney recipients from the DIVAT cohort alive with a functioning graft and a surveillance biopsy at 1-year post-transplantation. Patients displaying abnormal histology (49%) (borderline, acute rejection, interstitial fibrosis and tubular atrophy [IFTA] grade 2 or 3, glomerulonephritis) were compared to the normal or subnormal (IFTA grade 1) histology group. Obtained from a lasso penalized logistic regression, the 1-RBI was composed of recipient gender, serum creatinine at 3, 6, and 12 month post-transplantation and anticlass II immunization at transplantation (internal validation: AUC = 0.71, 95% CI [0.53-0.83]; external validation: AUC = 0.62, 95% CI [0.58-0.66]). While we could not determinate a threshold able to identify patients at high chance of normal or subnormal histology, we estimated and validated a discriminating threshold capable of identifying a subgroup of 15% of the patients with a risk of abnormal histology higher than 80%. The 1-RBI is computable online at www.divat.fr. The 1-RBI could be a useful tool to standardize 1-year biopsy proposal and may for instance help to indicate one in case of high risk of abnormal histology.
Collapse
Affiliation(s)
- Magali Giral
- CRTI UMR 1064, Inserm, Université de Nantes, ITUN, CHU Nantes, RTRS Centaure, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Labex Transplantex, Nantes, France
| | - Karine Renaudin
- Pathological Anatomy and Cytology, CHU Hôtel-Dieu, Nantes, France
| | - Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Redmer Luning
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Dany Anglicheau
- Kidney Transplant Center, Necker University Hospital, APHP, RTRS "Centaure", Paris Descartes and Sorbonne Paris Cité Universities, Paris, France
| | - Emmanuel Morelon
- Nephrology, Transplantation and Clinic Immunology Department, RTRS "Centaure", Edouard Herriot University Hospital, Hospices Civils, Lyon, France
| | - Alexandre Huneau
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
| | - Chloé Paul
- CRTI UMR 1064, Inserm, Université de Nantes, ITUN, CHU Nantes, RTRS Centaure, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Labex Transplantex, Nantes, France
| | - Sophie Brouard
- CRTI UMR 1064, Inserm, Université de Nantes, ITUN, CHU Nantes, RTRS Centaure, Nantes, France
- Centre d'Investigation Clinique en Biothérapie, Labex Transplantex, Nantes, France
| | - Grégoire Couvrat-Desvergnes
- CRTI UMR 1064, Inserm, Université de Nantes, ITUN, CHU Nantes, RTRS Centaure, Nantes, France
- Department of Nephrology, Dialysis and Transplantation, Departmental Hospital of Vendée, La Roche-sur-Yon, France
| | - Yohann Foucher
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
- CHU NANTES, Nantes, France
| | - Etienne Dantan
- INSERM UMR 1246 - SPHERE, Nantes University, Tours University, Nantes, France
| |
Collapse
|
169
|
Severova-Andreevska G, Grcevska L, Petrushevska G, Cakalaroski K, Sikole A, Stojceva–Taneva O, Danilovska I, Ivanovski N. The Spectrum of Histopathological Changes in the Renal Allograft - a 12 Months Protocol Biopsy Study. Open Access Maced J Med Sci 2018; 6:606-612. [PMID: 29731924 PMCID: PMC5927487 DOI: 10.3889/oamjms.2018.162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/16/2017] [Accepted: 03/17/2018] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Renal transplantation became a routine and successful medical treatment for Chronic Kidney Disease in the last 30 years all over the world. Introduction of Luminex based Single Antigen Beads (SAB) and recent BANFF consensus of histopathological phenotypes of different forms of rejection enables more precise diagnosis and changes the therapeutic approach. The graft biopsies, protocol or cause, indicated, remain a golden diagnostic tool for clinical follow up of kidney transplant recipients (KTR). AIM The study aimed to analyse the histopathological changes in renal grafts 12 months after the surgery in KTR with satisfactory kidney function. MATERIAL AND METHODS A 12-month protocol biopsy study was performed in a cohort of 50 Kidney transplant recipients (42 from living and 8 from deceased donors). Usual work-up for suitable donors and recipients, standard surgical procedure, basic principles of peri and postoperative care and follow up were done in all KTR. Sequential quadruple immunosuppression including induction with Anti-thymocyte globulin (ATG) or Interleukin-2R antagonist (IL-2R), and triple drug maintenance therapy with Calcineurin Inhibitors (CNI), Mycophenolate Mofetil (MMF) and Steroids were prescribed to all pts. Different forms of Glomerulonephritis (16), Hypertension (10), End Stage Renal Disease (13), Hereditary Nephropathies (6), Diabetes (3) and Vesicoureteral Reflux (2) were the underlying diseases. All biopsies were performed under ultrasound guidance. The 16 gauge needles with automated "gun" were used to take 2 cores of tissue. The samples were stained with HE, PAS, Trichrome Masson and Silver and reviewed by the same pathologist. A revised and uploaded BANFF 2013 classification in 6 categories (Cat) was used. RESULTS Out of 48 biopsies, 15 (31%) were considered as normal, 4 (8%), Borderline (BL-Cat 3), 5 (10%) as Interstitial Fibrosis/Tubular Atrophy (IF/TA-Cat 5), 5 (10%) were classified as non-immunological (Cat 6), 2 as a pure antibody-mediated rejection (ABMR-Cat 2) and T-cell Mediated Rejection (TCMR-Cat 4). The remaining 17 samples were classified as a "mixed" rejection: 7 (41%) ABMR + IF/TA, 5 (29%) ABMR + BL + IF/TA, 2 (11%) BL + IF/TA, 1 (5%) ABMR + BL, 1 (5%) ABMR + TCMR and 1 (5%) TCMR + IF/TA. The mean serum creatinine at the time of the biopsy was 126.7 ± 23.4 µmol/L, while GFR-MDRD 63.4 ± 20.7 ml/min, which means that the majority of the findings were subclinical. Among the non-immunological histological findings (Cat 6), 3 cases belonged to CNI toxicity, 1 to BK nephropathy and 1 to recurrence of the primary disease. CONCLUSION Our 12-month protocol biopsy study revealed the presence of different forms of mixed subclinical rejection. Use of recent BANFF classification and scoring system enables more precise diagnosis and subsequently different approach to the further treatment of the KTR. More correlative long-term studies including Anti HLA antibodies and Endothelial Cell Activation- Associated Transcripts (ENDAT) are needed.
Collapse
Affiliation(s)
- Galina Severova-Andreevska
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Ladislava Grcevska
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Gordana Petrushevska
- Institute for Pathology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Koco Cakalaroski
- Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Aleksandar Sikole
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Olivera Stojceva–Taneva
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Ilina Danilovska
- University Clinic of Nephrology, Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| | - Ninoslav Ivanovski
- Medical Faculty, University St Cyril and Methodius of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
170
|
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: 125] [Impact Index Per Article: 17.9] [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
|
171
|
Yamamoto T, Iwasaki K, Murotani K, Takeda A, Futamura K, Okada M, Tsujita M, Hiramitsu T, Goto N, Narumi S, Watarai Y, Morozumi K, Uchida K, Kobayashi T. Peripheral blood immune response-related gene analysis for evaluating the potential risk of chronic antibody-mediated rejection. Hum Immunol 2018; 79:432-438. [PMID: 29614336 DOI: 10.1016/j.humimm.2018.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/24/2018] [Accepted: 03/28/2018] [Indexed: 12/15/2022]
Abstract
Noninvasive methods for the early diagnosis of chronic antibody-mediated rejection (cAMR) are desired for patients with de novo (dn) donor-specific HLA antibody (DSA). This study aimed to elucidate the clinical relevance of immune-related gene expression in peripheral blood of kidney transplant recipients. The expression levels of fourteen key molecules (Foxp3, CTLA-4, CCR7, TGF-β, IGLL-1, IL-10, ITCH, CBLB, Bcl-6, CXCR5, granzyme B, CIITA, Baff, TOAG-1/TCAIM) related to regulatory/cytotoxic function of immune cells were compared in 93 patients, which were divided into Groups A (clinical cAMR with dn DSA, n = 16), B (subclinical cAMR with dn DSA, n = 17), C (negative cAMR with dn DSA, n = 21) and D (stable function without dn DSA, n = 39). CIITA mRNA expression levels in groups B and C were significantly lower than those in group D (p < 0.01). Moreover, the CTLA-4 mRNA expression in group A was significantly higher than that in groups B and C (p < 0.01). ROC curve analysis suggested that CIITA (AUC = 0.902) and CTLA-4 (AUC = 0.785) may serve as valuable biomarkers of the stage of dn DSA production and clinical cAMR, respectively. In addition to dn DSA screening, monitoring of CIITA and CTLA-4 in peripheral blood could offer useful information on the time course of the development of cAMR.
Collapse
Affiliation(s)
- Takayuki Yamamoto
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan; Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, AL, USA
| | - Kenta Iwasaki
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenta Murotani
- Division of Biostatistics, Clinical Research Center, Aichi Medical University Hospital, Nagakute, Japan
| | - Asami Takeda
- Department of Nephrology, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Kenta Futamura
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Manabu Okada
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Makoto Tsujita
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Takahisa Hiramitsu
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Norihiko Goto
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Shunji Narumi
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Yoshihiko Watarai
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Kunio Morozumi
- Department of Nephrology, Masuko Memorial Hospital, Nagoya, Japan
| | - Kazuharu Uchida
- Department of Kidney Disease and Transplant Immunology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi Medical University School of Medicine, Nagakute, Japan.
| |
Collapse
|
172
|
Redondo-Pachón D, Pérez-Sáez MJ, Mir M, Gimeno J, Llinás L, García C, Hernández JJ, Yélamos J, Pascual J, Crespo M. Impact of persistent and cleared preformed HLA DSA on kidney transplant outcomes. Hum Immunol 2018. [PMID: 29524568 DOI: 10.1016/j.humimm.2018.02.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Preformed HLA donor-specific antibodies (DSA) only detected with Luminex have been associated with increased risk of antibody-mediated rejection (ABMR) and graft failure after kidney transplantation (KT). Their evolution after KT may modify this risk. We analyzed postransplant evolution of preformed DSA identified retrospectively and their impact on outcomes of 370 KT performed 2006-2014. Antibodies were monitored prospectively at 1-3-5 years after KT and if any dysfunction. Early acute ABMR was more frequent among patients with preformed DSA class-I or I + II than isolated class-II (29.4% vs 4.5%, p = 0.02). One year post-KT, 20 of 34 patients with functioning KT had persistent DSA. Preformed DSA class-II persisted more frequently than class-I/I + II (66.7% vs 33.3%; p = 0.031). The only risk factor independently associated with persistence was pretransplant MFI. Patients with de novo DSA had the highest risk of ABMR (HR 22.2 [CI 6.1-81.2]). Although recipients with persisting preformed DSA had significantly increased ABMR risk (HR 14.7 [CI 6.5-33.0]), those with cleared preformed DSA also had a higher risk than those without DSA (HR 7.01 [CI 2.2-21.8]). Preformed DSA are a very important risk factor for ABMR and graft loss. Patients who clear preformed DSA still show an increased risk of ABMR and graft loss after KT.
Collapse
Affiliation(s)
- Dolores Redondo-Pachón
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain
| | - María José Pérez-Sáez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain
| | - Marisa Mir
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain
| | - Javier Gimeno
- Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain; Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Laura Llinás
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain
| | - Carmen García
- Laboratori de Referencia de Catalunya, Barcelona, Spain
| | | | - Jose Yélamos
- Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain; Department of Immunology, Hospital del Mar, Barcelona, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain.
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; Institute Mar for Medical Research, Parc de Salut Mar, Barcelona, Spain.
| |
Collapse
|
173
|
Early Conversion to Belatacept in Kidney Transplant Recipients With Low Glomerular Filtration Rate. Transplantation 2018; 102:478-483. [DOI: 10.1097/tp.0000000000001985] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
174
|
Zhu N, Rowe NE, Martin PR, Luke SS, Mcgregor TB, Myslik F, Mcalister VC, Sener A, Luke PP. Long-term results of protocol kidney biopsy directing steroid withdrawal in simultaneous pancreas-kidney transplant patients. Can Urol Assoc J 2018; 12:188-192. [PMID: 29485032 DOI: 10.5489/cuaj.4702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION We sought to determine whether protocol biopsies could be used to guide treatment and improve outcomes in simultaneous pancreas-kidney (SPK) patients. METHODS Between 2004 and 2013, protocol biopsies were performed on SPK patients at 3-6 months and one year post-transplant. Maintenance immunosuppression consisted of a calcineurin inhibitor, anti-proliferative agent, and corticosteroid. Corticosteroid was withdrawn in negative early biopsies, maintained in subclinical/ borderline biopsies, and increased if Banff IB or greater rejection was identified. Endpoints included presence of interstitial fibrosis and tubular atrophy on biopsy at one year (IF/TA), rejection episodes, and renal and pancreas function at five years' followup. RESULTS Forty-one SPK transplant patients were reviewed and a total of 75 protocol biopsies were identified. On early biopsy, 51% had negative biopsies, 44% had borderline rejection, and 5% had subclinical rejection. Renal and pancreas function were not significantly different at one, two, and five years post-transplant between negative vs. borderline early biopsy patients. No difference in the degree of IF/TA was found between these two groups. CONCLUSIONS To our knowledge, this is the first study to evaluate protocol biopsies as an investigative tool prior to steroid withdrawal in SPK patients. Our study suggests that there are no detrimental functional or histological effects at five years post-transplant, despite weaning steroids in the negative biopsy group.
Collapse
Affiliation(s)
- Nemin Zhu
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada
| | - Neal E Rowe
- Department of Surgery, University of Ottawa, Ottawa, ON; Canada
| | - Paul R Martin
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada.,Department of Surgery, London Health Sciences Centre, Western University, London, ON; Canada
| | - Sean S Luke
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada
| | | | - Frank Myslik
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada
| | - Vivian C Mcalister
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada.,Department of Surgery, London Health Sciences Centre, Western University, London, ON; Canada.,Multi-Organ Transplant Program, London Health Sciences Centre, London, ON; Canada
| | - Alp Sener
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada.,Department of Surgery, London Health Sciences Centre, Western University, London, ON; Canada.,Multi-Organ Transplant Program, London Health Sciences Centre, London, ON; Canada
| | - Patrick P Luke
- Western University Schulich School of Medicine and Dentistry, London, ON; Canada.,Department of Surgery, London Health Sciences Centre, Western University, London, ON; Canada.,Multi-Organ Transplant Program, London Health Sciences Centre, London, ON; Canada
| |
Collapse
|
175
|
de Leur K, Clahsen-van Groningen MC, van den Bosch TPP, de Graav GN, Hesselink DA, Samsom JN, Baan CC, Boer K. Characterization of ectopic lymphoid structures in different types of acute renal allograft rejection. Clin Exp Immunol 2018; 192:224-232. [PMID: 29319177 DOI: 10.1111/cei.13099] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 12/15/2022] Open
Abstract
We hypothesize that T cells such as interleukin (IL)-21+ B cell lymphoma 6 (BCL6)+ T follicular helper cells can regulate B cell-mediated immunity within the allograft during acute T cell-mediated rejection; this process may feed chronic allograft rejection in the long term. To investigate this mechanism, we determined the presence and activation status of organized T and B cells in so-called ectopic lymphoid structures (ELSs) in different types of acute renal allograft rejection. Biopsies showing the following primary diagnosis were included: acute/active antibody-mediated rejection, C4d+ (a/aABMR), acute T cell-mediated rejection grade I (aTCMRI) and acute T cell-mediated rejection grade II (aTCMRII). Paraffin sections were stained for T cells (CD3 and CD4), B cells (CD20), follicular dendritic cells (FDCs, CD23), activated B cells (CD79A), immunoglobulin (Ig)D, cell proliferation (Ki67) and double immunofluorescent stainings for IL-21 and BCL6 were performed. Infiltrates of T cells were detected in all biopsies. In aTCMRI, B cells formed aggregates surrounded by T cells. In these aggregates, FDCs, IgD and Ki67 were detected, suggesting the presence of ELSs. In contrast, a/aABMR and aTCMRII showed diffuse infiltrates of T and B cells but no FDCs and IgD. IL-21 was present in all biopsies. However, co-localization with BCL6 was observed mainly in aTCMRI biopsies. In conclusion, ELSs with an activated phenotype are found predominantly in aTCMRI where T cells co-localize with B cells. These findings suggest a direct pathway of B cell alloactivation at the graft site during T cell mediated rejection.
Collapse
Affiliation(s)
- K de Leur
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.,Division of HPB and Transplant Surgery, Department of Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - T P P van den Bosch
- Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - G N de Graav
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - D A Hesselink
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C C Baan
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - K Boer
- Section Transplantation and Nephrology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
176
|
Biomarker Guidelines for High-Dimensional Genomic Studies in Transplantation: Adding Method to the Madness. Transplantation 2018; 101:457-463. [PMID: 28212255 DOI: 10.1097/tp.0000000000001622] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
177
|
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
|
178
|
Lefaucheur C, Gosset C, Rabant M, Viglietti D, Verine J, Aubert O, Louis K, Glotz D, Legendre C, Duong Van Huyen JP, Loupy A. T cell-mediated rejection is a major determinant of inflammation in scarred areas in kidney allografts. Am J Transplant 2018; 18:377-390. [PMID: 29086461 DOI: 10.1111/ajt.14565] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/22/2017] [Accepted: 10/22/2017] [Indexed: 01/25/2023]
Abstract
Inflammation in fibrosis areas (i-IF/TA) of kidney allografts is associated with allograft loss; however, its diagnostic significance remains to be determined. We investigated the clinicohistologic phenotype and determinants of i-IF/TA in a prospective cohort of 1539 kidney recipients undergoing evaluation of i-IF/TA and tubulitis in atrophic tubules (t-IF/TA) on protocol allograft biopsies performed at 1 year posttransplantation. We considered donor, recipient, and transplant characteristics, immunosuppression, and histological diagnoses in 2260 indication biopsies performed within the first year posttransplantation. Nine hundred forty-six (61.5%) patients presented interstitial fibrosis/tubular atrophy (IF/TA Banff grade > 0) at 1 year posttransplant, among whom 394 (41.6%) showed i-IF/TA. i-IF/TA correlated with concurrent t-IF/TA (P < .001), interstitial inflammation (P < .001), tubulitis (P < .001), total inflammation (P < .001), peritubular capillaritis (P < .001), interstitial fibrosis (P < .001), and tubular atrophy (P = .02). The independent determinants of i-IF/TA were previous T cell-mediated rejection (TCMR) (P < .001), BK virus nephropathy (P = .007), steroid therapy (P = .039), calcineurin inhibitor therapy (P = .011), inosine-5'-monophosphate dehydrogenase inhibitor therapy (P = .011), HLA-B mismatches (P = .012), and HLA-DR mismatches (P = .044). TCMR patients with i-IF/TA on posttreatment biopsy (N = 83/136, 61.0%) exhibited accelerated progression of IF/TA over time (P = .01) and decreased 8-year allograft survival (70.8% vs 83.5%, P = .038) compared to those without posttreatment i-IF/TA. Our results support that i-IF/TA may represent a manifestation of chronic active TCMR.
Collapse
Affiliation(s)
- Carmen Lefaucheur
- Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Clément Gosset
- Department of Pathology, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Marion Rabant
- Department of Pathology, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Denis Viglietti
- Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Jérôme Verine
- Department of Pathology, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Olivier Aubert
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Kevin Louis
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Denis Glotz
- Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.,Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France
| | - Christophe Legendre
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-Paul Duong Van Huyen
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Pathology, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, Paris, France.,Department of Kidney Transplantation, Necker Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
179
|
Abstract
Donor-specific antibodies have become an established biomarker predicting antibody-mediated rejection. Antibody-mediated rejection is the leading cause of graft loss after kidney transplant. There are several phenotypes of antibody-mediated rejection along post-transplant course that are determined by the timing and extent of humoral response and the various characteristics of donor-specific antibodies, such as antigen classes, specificity, antibody strength, IgG subclasses, and complement binding capacity. Preformed donor-specific antibodies in sensitized patients can trigger hyperacute rejection, accelerated acute rejection, and early acute antibody-mediated rejection. De novo donor-specific antibodies are associated with late acute antibody-mediated rejection, chronic antibody-mediated rejection, and transplant glomerulopathy. The pathogeneses of antibody-mediated rejection include not only complement-dependent cytotoxicity, but also complement-independent pathways of antibody-mediated cellular cytotoxicity and direct endothelial activation and proliferation. The novel assay for complement binding capacity has improved our ability to predict antibody-mediated rejection phenotypes. C1q binding donor-specific antibodies are closely associated with acute antibody-mediated rejection, more severe graft injuries, and early graft failure, whereas C1q nonbinding donor-specific antibodies correlate with subclinical or chronic antibody-mediated rejection and late graft loss. IgG subclasses have various abilities to activate complement and recruit effector cells through the Fc receptor. Complement binding IgG3 donor-specific antibodies are frequently associated with acute antibody-mediated rejection and severe graft injury, whereas noncomplement binding IgG4 donor-specific antibodies are more correlated with subclinical or chronic antibody-mediated rejection and transplant glomerulopathy. Our in-depth knowledge of complex characteristics of donor-specific antibodies can stratify the patient's immunologic risk, can predict distinct phenotypes of antibody-mediated rejection, and hopefully, will guide our clinical practice to improve the transplant outcomes.
Collapse
Affiliation(s)
- Rubin Zhang
- Section of Nephrology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
180
|
Montgomery RA, Loupy A, Segev DL. Antibody-mediated rejection: New approaches in prevention and management. Am J Transplant 2018; 18 Suppl 3:3-17. [PMID: 29292861 DOI: 10.1111/ajt.14584] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/26/2017] [Accepted: 11/04/2017] [Indexed: 01/25/2023]
Abstract
Despite the success of desensitization protocols, antibody-mediated rejection (AMR) remains a significant contributor to renal allograft failure in patients with donor-specific antibodies. Plasmapheresis and high-dose intravenous immunoglobulin have proved to be effective treatments to prevent and treat AMR, but irreversible injury in the form of transplant glomerulopathy can commonly manifest months to years later. There is an unmet need to improve the outcomes for patients at risk for AMR. Updated Banff criteria now take into account the increasing understanding of the complex and heterogeneous nature of AMR phenotypes, including the timing of rejection, subclinical and chronic AMR, C4d-negative AMR, and antibody-mediated vascular rejection. Treatment for AMR is not standardized, and there is little in the way of evidence-based treatment guidelines. Refining more precisely the mechanisms of injury responsible for different AMR phenotypes and establishing relevant surrogate endpoints to facilitate more informative studies will likely allow for more accurate determination of prognosis and efficacious intervention using new therapeutic approaches. In addition to plasma exchange and intravenous immunoglobulin, a number of other add-on therapies have been tried in small studies without consistent benefit, including anti-CD20, proteasome inhibitors, complement inhibitors, anti-interleukin-6 receptor blockers, and immunoglobulin G-degrading enzyme of Streptococcus pyogenes (called IdeS).
Collapse
Affiliation(s)
- R A Montgomery
- Department of Surgery and NYU Langone Transplant Institute, NYU Langone Medical Center, New York, NY, USA
| | - A Loupy
- Paris Translational Research Center for Organ Transplantation and Department of Nephrology and Kidney Transplantation, Hôpital Necker, INSERM U 970, Paris Descartes University, Paris, France
| | - D L Segev
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
181
|
Parajuli S, Reville PK, Ellis TM, Djamali A, Mandelbrot DA. Utility of protocol kidney biopsies for de novo donor-specific antibodies. Am J Transplant 2017; 17:3210-3218. [PMID: 28805293 DOI: 10.1111/ajt.14466] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/24/2017] [Accepted: 07/29/2017] [Indexed: 01/25/2023]
Abstract
There is limited information about the role of protocol kidney biopsies for de novo donor-specific antibodies (dnDSA) in kidney transplant recipients, especially in those with stable graft function. We initiated a routine posttransplant DSA monitoring and surveillance biopsy program for dnDSA since 2014. We identified 45 kidney transplant recipients with dnDSA detected between January 2014 and February 2017 who underwent kidney biopsy within 60 days of detection of dnDSA. Twenty-nine (64%) had stable graft function and 16 (36%) had impaired graft function at the time of dnDSA detection. Even in the group with stable graft function, we found a high rate of rejection (53%) on biopsy. Eighty-eight percent of patients with impaired graft function had rejection. Those patients with impaired graft function had significantly lower estimated glomerular filtration rate at 12 months postbiopsy and at last follow-up. Those with impaired graft function had more graft failures; however, this result was not statistically significant. The high rate of asymptomatic rejection, and the fact that outcomes in asymptomatic patients are poor, is in support of the utility of surveillance biopsies in patients with dnDSA.
Collapse
Affiliation(s)
- Sandesh Parajuli
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Patrick K Reville
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Thomas M Ellis
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Arjang Djamali
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Didier A Mandelbrot
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
182
|
Steggerda JA, Kim IK, Haas M, Zhang X, Kang A, Pizzo H, Kamil E, Jordan S, Puliyanda D. Clinical and histopathologic features of antibody-mediated rejection among pediatric renal transplant recipients with preformed vs de novo donor-specific antibodies. Pediatr Transplant 2017; 21. [PMID: 29159992 DOI: 10.1111/petr.13079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2017] [Indexed: 01/18/2023]
Abstract
Preformed and de novo donor specific antibodies (pDSA and dnDSA) are risk factors for ABMR. This study compares the effects of pDSA vs dnDSA in pediatric kidney transplant recipients. Sixteen pediatric patients with biopsy-proven ABMR were evaluated. Strong DSA (MFI >10 000) was recorded at transplant, rejection, and follow-up. DSAs with the highest MFI were termed iDSAs. Allograft biopsies were scored according to Banff 2013 criteria. Seven of 16 (44%) patients had pDSA at transplant; 9 (56%) developed dnDSA. Patients with pDSA developed ABMR earlier (median = 63 vs 1344 days, P = .017), while patients with dnDSA were more likely to have strong Class II iDSA (100% vs 28%, P = .009). Viral infection or non-adherence was more common in patients developing dnDSA (88.8% vs 28.6%, P < .01). Pathology in those with pDSAs demonstrated worse transplant glomerulitis (g score 1.57 ± 0.98 vs 0.56 ± 0.73, P = .031); however, those with dnDSAs exhibited higher C4d+ ABMR (P = .013). Patients developing dnDSAs showed ABMR later post-transplant with predominance of HLA-Class II iDSAs. Inadequate immunosuppression likely contributes to dnDSA formation. Patients with no DSA who have unprotocolized decreases in immunosuppression should be screened for dnDSA as it could lead to early intervention and potentially better outcomes.
Collapse
Affiliation(s)
- Justin A Steggerda
- Division of Transplantation, Department of General Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Irene K Kim
- Division of Transplantation, Department of General Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mark Haas
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaohai Zhang
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexis Kang
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Helen Pizzo
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elaine Kamil
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stanley Jordan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dechu Puliyanda
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
183
|
Evidence for an important role of both complement-binding and noncomplement-binding donor-specific antibodies in renal transplantation. Curr Opin Organ Transplant 2017; 21:433-40. [PMID: 27348472 DOI: 10.1097/mot.0000000000000324] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW The review describes the current clinical relevance of circulating anti-human leukocyte antigen (anti-HLA) antibodies in kidney transplantation and discusses recent improvements in their characterization that provide new insights into the identification and management of important clinical outcomes. RECENT FINDINGS Recent studies addressing the relationships between donor-specific anti-HLA antibody (HLA-DSA) properties (i.e., their strength, complement-binding capacity, and IgG subclass composition) and allograft injury and survival have highlighted their relevance in the prediction of antibody-mediated injury and allograft loss. SUMMARY Antibody-mediated rejection is the leading cause of kidney allograft loss. Although considerable experimental and clinical evidence suggests a causal effect of circulating HLA-DSAs in antibody-mediated rejection and allograft failure, HLA-DSAs induce a wide spectrum of injuries to the allograft that illustrate the need to delineate the characteristics of HLA-DSAs that confer pathogenesis. Current risk stratification is based on HLA-DSA characteristics, including antibody specificity, HLA class, and strength. Recently, the complement-binding capacity of HLA-DSAs has been recognized as a clinically relevant marker for predicting pathogenicity and allograft loss. Emerging data also support a role for HLA-DSA IgG subclass composition in discriminating distinct patterns of antibody-mediated injury. This progress in our understanding of HLA-DSA pathogenicity provides new tools to stratify individual immunological risks. However, specific prospective studies addressing immunological risk stratification in large and unselected populations are required to define the clinical benefit and cost-effectiveness of such a comprehensive assessment of HLA-DSAs before implementation in current clinical practice.
Collapse
|
184
|
Abstract
Purpose of review Accurate and timely detection and characterization of human leukocyte antigen (HLA) antibodies are critical for pre-transplant and post-transplant immunological risk assessment. Solid phase immunoassays have provided increased sensitivity and specificity, but test interpretation is not always straightforward. This review will discuss the result interpretation considering technical limitations; assessment of relative antibody strength; and the integration of data for risk stratification from complementary testing and the patient's immunological history. Recent findings Laboratory and clinical studies have provided insight into causes of test failures – false positive reactions because of antibodies to denatured HLA antigens and false negative reactions resulting from test interference and/or loss of native epitopes. Test modifications permit detection of complement-binding antibodies and determination of the IgG subclasses. The high degree of specificity of single antigen solid phase immunoassays has revealed the complexity and clinical relevance of antibodies to HLA-C, HLA-DQ, and HLA-DP antigens. Determination of antibody specificity for HLA epitopes enables identification of incompatible antigens not included in test kits. Summary Detection and characterization of HLA antibodies with solid phase immunoassays has led to increased understanding of the role of those antibodies in graft rejection, improved treatment of antibody-mediated rejection, and increased opportunities for transplantation. However, realization of these benefits requires careful and accurate interpretation of test results.
Collapse
|
185
|
Risk factors associated with the development of histocompatibility leukocyte antigen sensitization. Curr Opin Organ Transplant 2017; 21:447-52. [PMID: 27258577 DOI: 10.1097/mot.0000000000000336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Despite excellent short-term kidney allograft survival rates, long-term outcomes have not improved. For years, the focus on improving these outcomes revolved around minimization or elimination of calcineurin toxicity. Despite our best efforts, approximately 5000 allografts are lost each year in the United States and results in a significant emotional burden for patients and financial burden for the healthcare system. RECENT FINDINGS Advancements in detection of donor-specific histocompatibility leukocyte antigen antibodies (DSAs) and improved assessment of allograft biopsy tissue have shown that the most common cause for graft failures is DSA-related antibody-mediated rejection. Sensitization is directly related to human tissue exposure prior to transplant. We now know that sensitization can occur in patients who are non compliant or poorly compliant with their calcineurin inhibitors. They develop de-novo DSAs, which are responsible for numerous allograft losses around the world. SUMMARY Given the current evidence, it is imperative that all transplant physicians recognize the importance of encouraging medication adherence to prevent the consequences of DSA-induced graft failure. However, little progress has been made in this area. Other potential therapeutic approaches based on B-cell depletion or modulation early posttransplant may help to reduce the risk for de-novo DSA development.
Collapse
|
186
|
Gleiss A, Oberbauer R, Heinze G. An unjustified benefit: immortal time bias in the analysis of time-dependent events. Transpl Int 2017; 31:125-130. [PMID: 29024071 DOI: 10.1111/tri.13081] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/03/2017] [Accepted: 10/06/2017] [Indexed: 11/27/2022]
Abstract
Immortal time bias is a problem arising from methodologically wrong analyses of time-dependent events in survival analyses. We illustrate the problem by analysis of a kidney transplantation study. Following patients from transplantation to death, groups defined by the occurrence or nonoccurrence of graft failure during follow-up seemingly had equal overall mortality. Such naive analysis assumes that patients were assigned to the two groups at time of transplantation, which actually are a consequence of occurrence of a time-dependent event later during follow-up. We introduce landmark analysis as the method of choice to avoid immortal time bias. Landmark analysis splits the follow-up time at a common, prespecified time point, the so-called landmark. Groups are then defined by time-dependent events having occurred before the landmark, and outcome events are only considered if occurring after the landmark. Landmark analysis can be easily implemented with common statistical software. In our kidney transplantation example, landmark analyses with landmarks set at 30 and 60 months clearly identified graft failure as a risk factor for overall mortality. We give further typical examples from transplantation research and discuss strengths and limitations of landmark analysis and other methods to address immortal time bias such as Cox regression with time-dependent covariables.
Collapse
Affiliation(s)
- Andreas Gleiss
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg Heinze
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
187
|
Adam B, Smith R, Rosales I, Matsunami M, Afzali B, Oura T, Cosimi A, Kawai T, Colvin R, Mengel M. Chronic Antibody-Mediated Rejection in Nonhuman Primate Renal Allografts: Validation of Human Histological and Molecular Phenotypes. Am J Transplant 2017; 17:2841-2850. [PMID: 28444814 PMCID: PMC5658276 DOI: 10.1111/ajt.14327] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/19/2017] [Accepted: 04/19/2017] [Indexed: 01/25/2023]
Abstract
Molecular testing represents a promising adjunct for the diagnosis of antibody-mediated rejection (AMR). Here, we apply a novel gene expression platform in sequential formalin-fixed paraffin-embedded samples from nonhuman primate (NHP) renal transplants. We analyzed 34 previously described gene transcripts related to AMR in humans in 197 archival NHP samples, including 102 from recipients that developed chronic AMR, 80 from recipients without AMR, and 15 normal native nephrectomies. Three endothelial genes (VWF, DARC, and CAV1), derived from 10-fold cross-validation receiver operating characteristic curve analysis, demonstrated excellent discrimination between AMR and non-AMR samples (area under the curve = 0.92). This three-gene set correlated with classic features of AMR, including glomerulitis, capillaritis, glomerulopathy, C4d deposition, and DSAs (r = 0.39-0.63, p < 0.001). Principal component analysis confirmed the association between three-gene set expression and AMR and highlighted the ambiguity of v lesions and ptc lesions between AMR and T cell-mediated rejection (TCMR). Elevated three-gene set expression corresponded with the development of immunopathological evidence of rejection and often preceded it. Many recipients demonstrated mixed AMR and TCMR, suggesting that this represents the natural pattern of rejection. These data provide NHP animal model validation of recent updates to the Banff classification including the assessment of molecular markers for diagnosing AMR.
Collapse
Affiliation(s)
- B.A. Adam
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - R.N. Smith
- Department of Pathology, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - I.A. Rosales
- Department of Pathology, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - M. Matsunami
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - B. Afzali
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - T. Oura
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - A.B. Cosimi
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - T. Kawai
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - R.B. Colvin
- Department of Pathology, Harvard Medical School and Massachusetts General Hospital, Boston, USA
| | - M. Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| |
Collapse
|
188
|
Kidney Transplant With Low Levels of DSA or Low Positive B-Flow Crossmatch: An Underappreciated Option for Highly Sensitized Transplant Candidates. Transplantation 2017; 101:2429-2439. [PMID: 28009780 DOI: 10.1097/tp.0000000000001619] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Avoiding donor-specific antibody (DSA) is difficult for sensitized patients. Improved understanding of the risk of low level DSA is needed. METHODS We retrospectively compared the outcomes of 954 patients transplanted with varied levels of baseline DSA detected by single antigen beads and B flow cytometric crossmatch (XM). Patients were grouped as follows: -DSA/-XM, +DSA/-XM, +DSA/low +XM, +DSA/high +XM, and -DSA/+XM and followed up for a mean of 4.1 ± 1.9 years (similar among groups, P = 0.49). RESULTS Death-censored allograft survival was similar in all groups except the +DSA/high +XM group, which was lower at 79.1% versus 96.2% in the -DSA/-XM group (P < 0.01). The incidence of chronic antibody-mediated rejection (CAMR) based on surveillance biopsy was higher with increasing DSA (8.2% -DSA/-XM, 17.0% +DSA/-XM, 30.6% +DSA/low +XM, and 51.2% +DSA/high +XM, P < 0.01), but similar in groups without baseline DSA (8.1% -DSA/-XM vs 15.4% -DSA/+XM, P = 0.19). Having a calculated panel-reactive antibody (cPRA) of 80% or greater was independently associated with CAMR (hazard ratio, 5.2; P = 0.03) even when DSA was undetected at baseline. By 2 years posttransplant, the incidence of CAMR was 19.4% in patients with cPRA of 80% or greater and undetected DSA and negative XM at baseline. CONCLUSIONS Kidney transplantation with low-level DSA with or without a low positive XM is a reasonable option for highly sensitized patients and may be advantageous compared with waiting for a negative XM deceased donor. The risk for CAMR is low in patients with no DSA even if the XM is positive. Patients with cPRA of 80% or greater are at risk for CAMR even if no DSA is detected.
Collapse
|
189
|
Van Loon E, Lerut E, Naesens M. The time dependency of renal allograft histology. Transpl Int 2017; 30:1081-1091. [DOI: 10.1111/tri.13042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/05/2017] [Accepted: 08/21/2017] [Indexed: 01/27/2023]
Affiliation(s)
- Elisabet Van Loon
- Laboratory of Nephrology; Department of Microbiology and Immunology; KU Leuven; Leuven Belgium
- Department of Nephrology and Renal Transplantation; University Hospitals Leuven; Leuven Belgium
| | - Evelyne Lerut
- Translational Cell and Tissue Research; Department of Imaging and Pathology; KU Leuven; Leuven Belgium
- Department of Morphology and Molecular Pathology; University Hospitals Leuven; Leuven Belgium
| | - Maarten Naesens
- Laboratory of Nephrology; Department of Microbiology and Immunology; KU Leuven; Leuven Belgium
- Department of Nephrology and Renal Transplantation; University Hospitals Leuven; Leuven Belgium
| |
Collapse
|
190
|
Molecular and Functional Noninvasive Immune Monitoring in the ESCAPE Study for Prediction of Subclinical Renal Allograft Rejection. Transplantation 2017; 101:1400-1409. [PMID: 27362314 DOI: 10.1097/tp.0000000000001287] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Subclinical acute rejection (sc-AR) is a main cause for functional decline and kidney graft loss and may only be assessed through surveillance biopsies. METHODS The predictive capacity of 2 novel noninvasive blood biomarkers, the transcriptional kidney Solid Organ Response Test (kSORT), and the IFN-γ enzyme-linked immunosorbent spot assay (ELISPOT) assay were assessed in the Evaluation of Sub-Clinical Acute rejection PrEdiction (ESCAPE) Study in 75 consecutive kidney transplants who received 6-month protocol biopsies. Both assays were run individually and in combination to optimize the use of these techniques to predict sc-AR risk. RESULTS Subclinical acute rejection was observed in 22 (29.3%) patients (17 T cell-mediated subclinical rejection [sc-TCMR], 5 antibody-mediated subclinical rejection [sc-ABMR]), whereas 53 (70.7%) showed a noninjured, preserved (stable [STA]) parenchyma. High-risk (HR), low-risk, and indeterminate-risk kSORT scores were observed in 15 (20%), 50 (66.7%), and 10 (13.3%) patients, respectively. The ELISPOT assay was positive in 31 (41%) and negative in 44 (58.7%) patients. The kSORT assay showed high accuracy predicting sc-AR (specificity, 98%; positive predictive value 93%) (all sc-ABMR and 58% sc-TCMR showed HR-kSORT), whereas the ELISPOT showed high precision ruling out sc-TCMR (specificity = 70%, negative predictive value = 92.5%), but could not predict sc-ABMR, unlike kSORT. The predictive probabilities for sc-AR, sc-TCMR, and sc-ABMR were significantly higher when combining both biomarkers (area under the curve > 0.85, P < 0.001) and independently predicted the risk of 6-month sc-AR in a multivariate regression analysis. CONCLUSIONS Combining a molecular and immune cell functional assay may help to identify HR patients for sc-AR, distinguishing between different driving alloimmune effector mechanisms.
Collapse
|
191
|
Impact of monocyte-macrophage inhibition by ibandronate on graft function and survival after kidney transplantation: a single-centre follow-up study over 15 years. Clin Exp Nephrol 2017; 22:474-480. [PMID: 28856476 DOI: 10.1007/s10157-017-1470-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/18/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the effect of ibandronate administration on long-term graft function and graft survival after successful renal transplantation. METHODS Seventy-two renal transplant recipients (36 patients each in the treatment and control group) were included and followed over a 15-year period. Data on graft function and death-censored transplant outcome were recorded at 1, 5, 10, and 15 years. RESULTS Death-censored Kaplan-Meier analysis showed significantly improved graft survival of the treatment group (p = 0.026), whereas Cox regression analysis showed that ibandronate was positively associated with improved transplant survival (p = 0.028, hazard ratio 0.24, 95% confidence interval 0.07-0.86). Although general linear modelling did not indicate that ibandronate had a significant effect on transplant function (calculated using the estimated glomerular filtration rate according to Chronic Kidney Disease Epidemiology Collaboration equation) over the entire 15-year period (p = 0.650), there was a tendency towards improved graft function 1-year post-transplantion (p = 0.056). CONCLUSIONS Ibandronate treatment within the first year of transplantation resulted in a trend towards better graft function within the first few year post-transplant, and was associated with increased transplant survival at long-term follow-up.
Collapse
|
192
|
Garg N, Samaniego MD, Clark D, Djamali A. Defining the phenotype of antibody-mediated rejection in kidney transplantation: Advances in diagnosis of antibody injury. Transplant Rev (Orlando) 2017; 31:257-267. [PMID: 28882367 DOI: 10.1016/j.trre.2017.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 08/08/2017] [Accepted: 08/10/2017] [Indexed: 11/29/2022]
Abstract
The diagnostic criteria for antibody-mediated rejection (ABMR) are constantly evolving in light of the evidence. Inclusion of C4d-negative ABMR has been one of the major advances in the Banff Classification in recent years. Currently Banff 2015 classification requires evidence of donor specific antibodies (DSA), interaction between DSA and the endothelium, and acute tissue injury (in the form of microvasculature injury (MVI); acute thrombotic microangiopathy; or acute tubular injury in the absence of other apparent cause). In this article we review not only the ABMR phenotypes acknowledged in the most recent Banff classification, but also the phenotypes related to novel pathogenic antibodies (non-HLA DSA, antibody isoforms and subclasses, complement-binding functionality) and molecular diagnostic tools (gene transcripts, metabolites, small proteins, cytokines, and donor-derived cell-free DNA). These novel tools are also being considered for the prognosis and monitoring of treatment response. We propose that improved classification of ABMR based on underlying pathogenic mechanisms and outcomes will be an important step in identifying patient-centered therapies to extend graft survival.
Collapse
Affiliation(s)
- Neetika Garg
- Department of Medicine, Nephrology Division, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, United States.
| | - Milagros D Samaniego
- Department of Medicine, Nephrology Division, University of Michigan, Ann Arbor, MI 48109, United States
| | - Dana Clark
- Department of Medicine, Nephrology Division, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, United States
| | - Arjang Djamali
- Department of Medicine, Nephrology Division, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, United States
| |
Collapse
|
193
|
Kurian S, Velazquez E, Thompson R, Whisenant T, Rose S, Riley N, Harrison F, Gelbart T, Friedewald J, charrette J, Brietigam S, Peysakhovich J, First M, Abecassis M, Salomon D. Orthogonal Comparison of Molecular Signatures of Kidney Transplants With Subclinical and Clinical Acute Rejection: Equivalent Performance Is Agnostic to Both Technology and Platform. Am J Transplant 2017; 17:2103-2116. [PMID: 28188669 PMCID: PMC5519433 DOI: 10.1111/ajt.14224] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/06/2017] [Accepted: 01/20/2017] [Indexed: 01/25/2023]
Abstract
We performed orthogonal technology comparisons of concurrent peripheral blood and biopsy tissue samples from 69 kidney transplant recipients who underwent comprehensive algorithm-driven clinical phenotyping. The sample cohort included patients with normal protocol biopsies and stable transplant (sTx) function (n = 25), subclinical acute rejection (subAR, n = 23), and clinical acute rejection (cAR, n = 21). Comparisons between microarray and RNA sequencing (RNA-seq) signatures were performed and demonstrated a strong correlation between the blood and tissue compartments for both technology platforms. A number of shared differentially expressed genes and pathways between subAR and cAR in both platforms strongly suggest that these two clinical phenotypes form a continuum of alloimmune activation. SubAR is associated with fewer or less expressed genes than cAR in blood, whereas in biopsy tissues, this clinical phenotype demonstrates a more robust molecular signature for both platforms. The discovery work done in this study confirms a clear ability to detect gene expression profiles for sTx, subAR, and cAR in both blood and biopsy tissue, yielding equivalent predictive performance that is agnostic to both technology and platform. Our data also provide strong biological insights into the molecular mechanisms underlying these signatures, underscoring their logistical potential as molecular diagnostics to improve clinical outcomes following kidney transplantation.
Collapse
Affiliation(s)
- S.M. Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - E. Velazquez
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - R. Thompson
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - T. Whisenant
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - S. Rose
- Transplant Genomics Inc., Mansfield, MA
| | - N. Riley
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - F. Harrison
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - T. Gelbart
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | - J.J. Friedewald
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - j. charrette
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - S. Brietigam
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - J. Peysakhovich
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - M.R. First
- Transplant Genomics Inc., Mansfield, MA,Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - M.M. Abecassis
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - D.R. Salomon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
194
|
Impact on mid-term kidney graft outcomes of pretransplant anti-HLA antibodies detected by solid-phase assays: Do donor-specific antibodies tell the whole story? Hum Immunol 2017; 78:526-533. [PMID: 28732720 DOI: 10.1016/j.humimm.2017.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 06/12/2017] [Accepted: 07/17/2017] [Indexed: 01/31/2023]
Abstract
The detrimental impact of preformed anti-HLA donor-specific antibodies (DSA) is well defined, contrarily to non-donor-specific antibodies (NDSA). We sought to evaluate their clinical impact in a cohort of 724 kidney graft recipients in whom anti-HLA antibodies were thoroughly screened and identified in pre-transplant sera by solid-phase assays. NDSA or DSA were detected in 100 (13.8%) and 47 (6.5%) recipients respectively, while 577 (79.7%) were non-allosensitized (NaS). Incidence of antibody-mediated rejection at 1-year was 0.7%, 4.0% and 25.5% in NaS, NDSA and DSA patients, respectively (NaS vs. NDSA P=0.004; NaS vs. DSA P<0.001; NDSA vs. DSA P<0.001). Graft survival was lowest in DSA (78.7%), followed by NDSA (88.0%) and NaS (93.8%) recipients (NaS vs. NDSA P=0.015; NaS vs. DSA P<0.001; NDSA vs. DSA P=0.378). Multivariable competing risk analysis confirmed both NDSA (sHR=2.19; P=0.025) and DSA (sHR=2.87; P=0.012) as significant predictors of graft failure. The negative effect of NDSA and DSA on graft survival was significant in patients receiving no induction (P=0.019) or an anti-IL-2 receptor antibody (P<0.001), but not in those receiving anti-thymocyte globulin (P=0.852). The recognition of the immunological risk associated with preformed DSA but also NDSA have important implications in patients' risk stratification, and may impact clinical decisions at transplant.
Collapse
|
195
|
Danger R, Sawitzki B, Brouard S. Immune monitoring in renal transplantation: The search for biomarkers. Eur J Immunol 2017; 46:2695-2704. [PMID: 27861809 DOI: 10.1002/eji.201545963] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 11/11/2022]
Abstract
It is now widely accepted that in order to improve long-term graft function and survival, a more personalized immunosuppressive treatment of transplant patients according to the individual anti-donor immune response status is needed. This applies to the identification of potentially "high-risk" patients likely to develop acute rejection episodes or display an accelerated decline of graft function, patients who might need immunosuppression intensification, and operationally tolerant patients suitable for immunosuppression minimization or weaning off. Such a patient stratification would benefit from biomarkers, which enable categorization into low and high risk or, ideally, identification of operational tolerant patients. Here, we report on recent developments regarding identification and performance analysis of noninvasive biomarkers such as mRNA and miRNA expression profiles, chemokines, or changes in immune cell subsets in either blood or urine of renal transplant patients. We will also discuss which future steps are needed to accelerate their clinical implementation.
Collapse
Affiliation(s)
- Richard Danger
- Inserm, , Center for Research in Transplantation and Immunology (CRTI) U1064, Nantes, France.,Université de Nantes, , UMR1064, Nantes, France.,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, France
| | - Birgit Sawitzki
- Institute of Medical Immunology, Charité University Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Berlin, Germany
| | - Sophie Brouard
- Inserm, , Center for Research in Transplantation and Immunology (CRTI) U1064, Nantes, France.,Université de Nantes, , UMR1064, Nantes, France.,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, France.,CIC Biotherapy, CHU Nantes, , 30 bd Jean-Monnet, Nantes, France
| |
Collapse
|
196
|
Sicard A, Meas-Yedid V, Rabeyrin M, Koenig A, Ducreux S, Dijoud F, Hervieu V, Badet L, Morelon E, Olivo-Marin JC, Dubois V, Thaunat O. Computer-assisted topological analysis of renal allograft inflammation adds to risk evaluation at diagnosis of humoral rejection. Kidney Int 2017; 92:214-226. [DOI: 10.1016/j.kint.2017.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 12/30/2016] [Accepted: 01/05/2017] [Indexed: 11/15/2022]
|
197
|
Valenzuela NM, Reed EF. Antibody-mediated rejection across solid organ transplants: manifestations, mechanisms, and therapies. J Clin Invest 2017; 127:2492-2504. [PMID: 28604384 DOI: 10.1172/jci90597] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Solid organ transplantation is a curative therapy for hundreds of thousands of patients with end-stage organ failure. However, long-term outcomes have not improved, and nearly half of transplant recipients will lose their allografts by 10 years after transplant. One of the major challenges facing clinical transplantation is antibody-mediated rejection (AMR) caused by anti-donor HLA antibodies. AMR is highly associated with graft loss, but unfortunately there are few efficacious therapies to prevent and reverse AMR. This Review describes the clinical and histological manifestations of AMR, and discusses the immunopathological mechanisms contributing to antibody-mediated allograft injury as well as current and emerging therapies.
Collapse
|
198
|
Inflammation in Early Kidney Allograft Surveillance Biopsies With and Without Associated Tubulointerstitial Chronic Damage as a Predictor of Fibrosis Progression and Development of De Novo Donor Specific Antibodies. Transplantation 2017; 101:1410-1415. [DOI: 10.1097/tp.0000000000001216] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
199
|
Abstract
Complement is a major contributor to inflammation and graft injury. This system is especially important in ischemia-reperfusion injury/delayed graft function as well as in acute and chronic antibody-mediated rejection (AMR). The latter is increasingly recognized as a major cause of late graft loss, for which we have few effective therapies. C1 inhibitor (C1-INH) regulates several pathways which contribute to both acute and chronic graft injuries. However, C1-INH spares the alternative pathway and the membrane attack complex (C5–9) so innate antibacterial defenses remain intact. Plasma-derived C1-INH has been used to treat hereditary angioedema for more than 30 years with excellent safety. Studies with C1-INH in transplant recipients are limited, but have not revealed any unique toxicity or serious adverse events attributed to the protein. Extensive data from animal and ex vivo models suggest that C1-INH ameliorates ischemia-reperfusion injury. Initial clinical studies suggest this effect may allow transplantation of donor organs which are now discarded because the risk of primary graft dysfunction is considered too great. Although the incidence of severe early AMR is declining, accumulating evidence strongly suggests that complement is an important mediator of chronic AMR, a major cause of late graft loss. Thus, C1-INH may also be helpful in preserving function of established grafts. Early clinical studies in transplantation suggest significant beneficial effects of C1-INH with minimal toxicity. Recent results encourage continued investigation of this already-available therapeutic agent.
Collapse
|
200
|
Abstract
Short-term outcomes in renal transplantation have improved significantly in the past few years. However, the improvement in long-term outcomes has been modest. The reasons for graft failure beyond the first year of transplantation have been attributed to several different factors. We believe that subclinical rejection (SCR) may be 1 of the factors that contribute to graft loss in the long run. We also believe that there are data to suggest that SCR leads to progressive fibrosis and loss of graft function. This has been demonstrated even in patients who have mild degrees of subclinical inflammation. This review outlines the major studies that have been published on this important topic. It also outlines potential risk factors for the development of SCR. The current approach and diagnostic methods are discussed as well as their pros and cons. Newer noninvasive methods of diagnosis as well as molecular diagnostics and their merits and shortcomings are also discussed in some depth. Thus, the proposed state of the art review on SCR will create a renewed interest at all levels including transplant clinicians, transplant researchers, pharmaceutical industries as well as regulatory organizations.
Collapse
|