دورية أكاديمية

The Histological Spectrum and Clinical Significance of T Cell-mediated Rejection of Kidney Allografts.

التفاصيل البيبلوغرافية
العنوان: The Histological Spectrum and Clinical Significance of T Cell-mediated Rejection of Kidney Allografts.
المؤلفون: Filippone EJ; Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA., Farber JL; Department of Pathology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA.
المصدر: Transplantation [Transplantation] 2023 May 01; Vol. 107 (5), pp. 1042-1055. Date of Electronic Publication: 2022 Dec 19.
نوع المنشور: Journal Article
اللغة: English
بيانات الدورية: Publisher: Lippincott Williams & Wilkins Country of Publication: United States NLM ID: 0132144 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1534-6080 (Electronic) Linking ISSN: 00411337 NLM ISO Abbreviation: Transplantation Subsets: MEDLINE
أسماء مطبوعة: Publication: Hagerstown, MD : Lippincott Williams & Wilkins
Original Publication: Baltimore, Williams & Wilkins.
مواضيع طبية MeSH: T-Lymphocytes* , Kidney Transplantation*/adverse effects, Humans ; Clinical Relevance ; Risk Factors ; Kidney/pathology ; Isoantibodies ; Inflammation/etiology ; Allografts/pathology ; Graft Rejection ; Biopsy
مستخلص: T cell-mediated rejection (TCMR) remains a significant cause of long-term kidney allograft loss, either indirectly through induction of donor-specific anti-HLA alloantibodies or directly through chronic active TCMR. Whether found by indication or protocol biopsy, Banff defined acute TCMR should be treated with antirejection therapy and maximized maintenance immunosuppression. Neither isolated interstitial inflammation in the absence of tubulitis nor isolated tubulitis in the absence of interstitial inflammation results in adverse outcomes, and neither requires antirejection treatment. RNA gene expression analysis of biopsy material may supplement conventional histology, especially in ambiguous cases. Lesser degrees of tubular and interstitial inflammation (Banff borderline) may portend adverse outcomes and should be treated when found on an indication biopsy. Borderline lesions on protocol biopsies may resolve spontaneously but require close follow-up if untreated. Following antirejection therapy of acute TCMR, surveillance protocol biopsies should be considered. Minimally invasive blood-borne assays (donor-derived cell-free DNA and gene expression profiling) are being increasingly studied as a means of following stable patients in lieu of biopsy. The clinical benefit and cost-effectiveness require confirmation in randomized controlled trials. Treatment of acute TCMR is not standardized but involves bolus corticosteroids with lymphocyte depleting antibodies for severe, refractory, or relapsing cases. Arteritis may be found with acute TCMR, active antibody-mediated rejection, or mixed rejections and should be treated accordingly. The optimal treatment ofchronic active TCMR is uncertain. Randomized controlled trials are necessary to optimally define therapy.
(Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
References: Hart A, Smith JM, Skeans MA, et al. OPTN/SRTR 2015 annual data report: kidney. Am J Transplant. 2017;17(Suppl 1):21–116.
Meier-Kriesche HU, Schold JD, Srinivas TR, et al. Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am J Transplant. 2004;4:378–383.
Sellarés J, de Freitas DG, Mengel M, et al. Understanding the causes of kidney transplant failure: the dominant role of antibody-mediated rejection and nonadherence. Am J Transplant. 2012;12:388–399.
Wiebe C, Gibson IW, Blydt-Hansen T, et al. Evolution and clinical pathologic correlations of de novo donor-specific HLA antibody post kidney transplant. Am J Transplant. 2012;12:1157–1167.
Solez K, Axelsen RA, Benediktsson H, et al. International standardization of criteria for the histologic diagnosis of renal allograft rejection: the Banff working classification of kidney transplant pathology. Kidney Int. 1993;44:411–422.
Racusen LC, Solez K, Colvin RB, et al. The Banff 97 working classification of renal allograft pathology. Kidney Int. 1999;55:713–723.
Racusen LC, Colvin RB, Solez K, et al. Antibody-mediated rejection criteria – an addition to the Banff ′97 classification of renal allograft rejection. Am J Transplant. 2003;3:708–714.
Solez K, Colvin RB, Racusen LC, et al. Banff ‘05 meeting report: differential diagnosis of chronic allograft injury and elimination of chronic allograft nephropathy (‘CAN’). Am J Transplant. 2007;7:518–526.
Solez K, Colvin RB, Racusen LC, et al. Banff 07 classification of renal allograft pathology: updates and future directions. Am J Transplant. 2008;8:753–760.
Sis B, Mengel M, Haas M, et al. Banff ’09 meeting report: antibody mediated graft deterioration and implementation of Banff working groups. Am J Transplant. 2010;10:464–471.
Mengel M, Sis B, Haas M, et al.; Banff Meeting Report Writing Committee. Banff 2011 meeting report: new concepts in antibody-mediated rejection. Am J Transplant. 2012;12:563–570.
Haas M, Sis B, Racusen LC, et al.; Banff Meeting Report Writing Committee. Banff 2013 meeting report: inclusion of c4d-negative antibody-mediated rejection and antibody-associated arterial lesions. Am J Transplant. 2014;14:272–283.
Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting report: current challenges in rejection classification and prospects for adopting molecular pathology. Am J Transplant. 2017;17:28–41.
Haas M, Loupy A, Lefaucheur C, et al. The Banff 2017 kidney meeting report: revised diagnostic criteria for chronic active T cell–mediated rejection, antibody-mediated rejection, and prospects for integrative endpoints for next-generation clinical trials. Am J Transplant. 2018;18:293–307.
Loupy A, Haas M, Roufosse C, et al. The Banff 2019 kidney meeting report (I): updates on and clarification of criteria for T cell– and antibody-mediated rejection. Am J Transplant. 2020;20:2318–2331.
Mengel M, Loupy A, Haas M, et al. Banff 2019 meeting report: molecular diagnostics in solid organ transplantation-consensus for the Banff human organ transplant (B-HOT) gene panel and open source multicenter validation. Am J Transplant. 2020;20:2305–2317.
Mueller TF, Einecke G, Reeve J, et al. Microarray analysis of rejection in human kidney transplants using pathogenesis-based transcript sets. Am J Transplant. 2007;7:2712–2722.
Madill-Thomsen KS, Wiggins RC, Eskandary F, et al. The effect of cortex/medulla proportions on molecular diagnoses in kidney transplant biopsies: Rejection and injury can be assessed in medulla. Am J Transplant. 2017;17:2117–2128.
Reeve J, Böhmig GA, Eskandary F, et al. Assessing rejection-related disease in kidney transplant biopsies based on archetypal analysis of molecular phenotypes. JCI Insight. 2017;2:e94197.
Reeve J, Böhmig GA, Eskandary F, et al.; INTERCOMEX MMDx-Kidney Study Group. Generating automated kidney transplant biopsy reports combining molecular measurements with ensembles of machine learning classifiers. Am J Transplant. 2019;19:2719–2731.
Madill-Thomsen K, Perkowska-Ptasińska A, Böhmig GA, et al. Discrepancy analysis comparing molecular and histology diagnoses in kidney transplant biopsies. Am J Transplant. 2020;20:1341–1350.
Smith RN. In-silico performance, validation, and modeling of the nanostring Banff human organ transplant gene panel using archival data from human kidney transplants. BMC Med Genomics. 2021;14:86.
van Baardwijk M, Cristoferi I, Ju J, et al. A decentralized kidney transplant biopsy classifier for transplant rejection developed using genes of the Banff-human organ transplant panel. Front Immunol. 2022;13:841519.
Filippone EJ, Farber JL. The monitoring of donor-derived cell-free DNA in kidney transplantation. Transplantation. 2021;105:509–516.
Xiao H, Gao F, Pang Q, et al. Diagnostic accuracy of donor-derived cell-free DNA in renal-allograft rejection: a meta-analysis. Transplantation. 2021;105:1303–1310.
Halloran PF, Reeve J, Madill-Thomsen KS, et al. Combining donor-derived cell-free DNA fraction and quantity to detect kidney transplant rejection using molecular diagnoses and histology as confirmation. Transplantation. 2022;106:2435–2442. doi:10.1097/TP.0000000000004212. (PMID: 10.1097/TP.0000000000004212)
Friedewald JJ, Kurian SM, Heilman RL, et al. 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.
Van Loon E, Gazut S, Yazdani S, et al. Development and validation of a peripheral blood mRNA assay for the assessment of antibody-mediated kidney allograft rejection: a multicentre, prospective study. EBioMedicine. 2019;46:463–472.
Zhang W, Yi Z, Keung KL, et al. A peripheral blood gene expression signature to diagnose subclinical acute rejection. J Am Soc Nephrol. 2019;30:1481.
Filippone EJ, Gulati R, Farber JL. Noninvasive assessment of the alloimmune response in kidney transplantation. Adv Chronic Kidney Dis. 2021;28:548–560.
Filippone EJ, Farber JL. The implications of B-lineage cells in kidney allografts. Transplantation. 2020;104:2011–2023.
Nankivell BJ, P’Ng CH, Chapman JR. Does tubulitis without interstitial inflammation represent borderline acute T cell mediated rejection? Am J Transplant. 2019;19:132–144.
Mehta RB, Tandukar S, Jorgensen D, et al. Early subclinical tubulitis and interstitial inflammation in kidney transplantation have adverse clinical implications. Kidney Int. 2020;98:436–447.
Mengel M, Gwinner W, Schwarz A, et al. Infiltrates in protocol biopsies from renal allografts. Am J Transplant. 2007;7:356–365.
Nankivell B, MD P, P’Ng C, et al. The pathophysiology and impact of inflammation in nonscarred renal interstitium: the Banff i lesion. Transplantation. 2020;104:835–846.
Ortiz F, Gelpi R, Helanterä I, et al. Decreased kidney graft survival in low immunological risk patients showing inflammation in normal protocol biopsies. PLoS One. 2016;11:e0159717.
Cosio FG, Grande JP, Wadei H, et al. Predicting subsequent decline in kidney allograft function from early surveillance biopsies. Am J Transplant. 2005;5:2464–2472.
Moreso F, Ibernon M, Gomà M, et al. Subclinical rejection associated with chronic allograft nephropathy in protocol biopsies as a risk factor for late graft loss. Am J Transplant. 2006;6:747–752.
Garcia-Carro C, Dorje C, Asberg A, et al. 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.
McRae M, Bouchard-Boivin F, Béland S, et al. Impact of the current versus the previous diagnostic threshold on the outcome of patients with borderline changes suspicious for T cell-mediated rejection diagnosed on indication biopsies. Transplantation. 2018;102:2120–2125.
Wiebe C, Rush DN, Gibson IW, et al. Evidence for the alloimmune basis and prognostic significance of borderline T cell-mediated rejection. Am J Transplant. 2020;20:2499–2508.
Rush DN, Jeffery JR, Gough J. Sequential protocol biopsies in renal transplant patients. clinico-pathological correlations using the Banff schema. Transplantation. 1995;59:511–514.
Schweitzer EJ, Drachenberg CB, Anderson L, et al. Significance of the Banff borderline biopsy. Am J Kidney Dis. 1996;28:585–588.
Gaber LW, Moore LW, Alloway RR, et al. Correlation between Banff classification, acute renal rejection scores and reversal of rejection. Kidney Int. 1996;49:481–487.
Serón D, Moreso F, Bover J, et al. Early protocol renal allograft biopsies and graft outcome. Kidney Int. 1997;51:310–316.
Saad R, Gritsch H, Shapiro R, et al. Clinical significance of renal allograft biopsies with “borderline changes,” as defined in the Banff schema 1. Transplantation. 1997;64:992–995.
Legendre C, Thervet E, Skhiri H, et al. Histologic features of chronic allograft nephropathy revealed by protocol biopsies in kidney transplant recipients. Transplantation. 1998;65:1506–1509.
Meehan SM, Siegel CT, Aronson AJ, et al. The relationship of untreated borderline infiltrates by the Banff criteria to acute rejection in renal allograft biopsies. J Am Soc Nephrol. 1999;10:1806–1814.
Lipman ML, Shen Y, Jeffery JR, et al. Immune-activation gene expression in clinically stable renal allograft biopsies: molecular evidence for subclinical rejection. Transplantation. 1998;66:1673–1681.
Bates WD, Davies DR, Welsh K, et al. An evaluation of the Banff classification of early renal allograft biopsies and correlation with outcome. Nephrol Dial Transplant. 1999;14:2364–2369.
Shapiro R, Randhawa P, Jordan ML, et al. An analysis of early renal transplant protocol biopsies--the high incidence of subclinical tubulitis. Am J Transplant. 2001;1:47–50.
Gloor J, Cohen A, Lager D, et al. Subclinical rejection in tacrolimus-treated renal transplant recipients. Transplantation. 2002;73:1965–1967.
Miyagi M, Ishikawa Y, Mizuiri S, et al. Significance of subclinical rejection in early renal allograft biopsies for chronic allograft dysfunction. Clin Transplant. 2005;19:456–465.
Choi BS, Shin MJ, Shin SJ, et al. Clinical significance of an early protocol biopsy in living-donor renal transplantation: ten-year experience at a single center. Am J Transplant. 2005;5:1354–1360.
Dahan K, Audard V, Roudot-Thoraval F, et al. Renal allograft biopsies with borderline changes: Predictive factors of clinical outcome. Am J Transplant. 2006;6:1725–1730.
Kee T, Chapman J, O’Connell P, et al. Treatment of subclinical rejection diagnosed by protocol biopsy of kidney transplants. Transplantation. 2006;82:36–42.
de Freitas DG, Sellarés J, Mengel M, et al. The nature of biopsies with “borderline rejection” and prospects for eliminating this category. Am J Transplant. 2012;12:191–201.
Wu K, Budde K, Lu H, et al. The severity of acute cellular rejection defined by Banff classification is associated with kidney allograft outcomes. Transplantation. 2014;97:1146–1154.
Hrubá P, Brabcová I, Gueler F, et al. Molecular diagnostics identifies risks for graft dysfunction despite borderline histologic changes. Kidney Int. 2015;88:785–795.
Zachariah MS, Dwivedi AK, Yip CS, et al. Utility of serial protocol biopsies performed after 1 year in predicting long-term kidney allograft function according to histologic phenotype. Exp Clin Transplant. 2018;16:391–400.
Mehta R, Bhusal S, Randhawa P, et al. 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.
Nankivell BJ, Agrawal N, Sharma A, et al. The clinical and pathological significance of borderline T cell-mediated rejection. Am J Transplant. 2019;19:1452–1463.
Lezoeva E, Nilsson J, Wüthrich R, et al. High PIRCHE scores may allow risk stratification of borderline rejection in kidney transplant recipients. Front Immunol. 2022;13:788818.
Fernández-Camargo DA, Marino L, Muñiz-Cuervo E, et al. Sub-classification of borderline changes into diffuse or focal and its impact on long-term renal transplant outcomes. Transpl Immunol. 2022;72:101594.
Beimler J, Zeier M. Borderline rejection after renal transplantation – to treat or not to treat. Clin Transplant. 2009;23:19–25.
Sorof JM, Vartanian RK, Olson JL, et al. Histopathological concordance of paired renal allograft biopsy cores. effect on the diagnosis and management of acute rejection. Transplantation. 1995;60:1215–1219.
Piovesan AC, Lucon AM, David DSR, et al. Multifocal renal allograft biopsy: Impact on therapeutic decisions. Transplant Proc. 2008;40:3397–3400.
Furness PN, Taub N. Convergence of European Renal Transplant Pathology Assessment Procedures (CERTPAP) Project. International variation in the interpretation of renal transplant biopsies: report of the CERTPAP Project. Kidney Int. 2001;60:1998–2012.
Gough J, Rush D, Jeffery J, et al. Reproducibility of the Banff schema in reporting protocol biopsies of stable renal allografts. Nephrol Dial Transplant. 2002;17:1081–1084.
Veronese FV, Manfro RC, Roman FR, et al. Reproducibility of the Banff classification in subclinical kidney transplant rejection. Clin Transplant. 2005;19:518–521.
Farris AB, Moghe I, Wu S, et al. Banff digital pathology working group: going digital in transplant pathology. Am J Transplant. 2020;20:2392–2399.
Reeve J, Einecke G, Mengel M, et al. Diagnosing rejection in renal transplants: a comparison of molecular- and histopathology-based approaches. Am J Transplant. 2009;9:1802–1810.
Mueller TF, Einecke G, Reeve J, et al. Microarray analysis of rejection in human kidney transplants using pathogenesis-based transcript sets. Am J Transplant. 2007;7:2712–2722.
Salazar ID, Merino López M, Chang J, et al. Reassessing the significance of intimal arteritis in kidney transplant biopsy specimens. J Am Soc Nephrol. 2015;26:3190–3198.
Wohlfahrtova M, Hruba P, Klema J, et al. Early isolated V-lesion may not truly represent rejection of the kidney allograft. Clin Sci (Lond). 2018;132:2269–2284.
Lefaucheur C, Loupy A, Vernerey D, et al. Antibody-mediated vascular rejection of kidney allografts: a population-based study. Lancet. 2013;381:313–319.
Nankivell BJ, Shingde M, P’Ng CH. The pathological and clinical diversity of acute vascular rejection in kidney transplantation. Transplantation. 2022;106:1666–1676.
Sis B, Bagnasco SM, Cornell LD, et al. Isolated endarteritis and kidney transplant survival: a multicenter collaborative study. J Am Soc Nephrol. 2015;26:1216.
Nankivell BJ, Borrows RJ, Fung CL, et al. Delta analysis of posttransplantation tubulointerstitial damage. Transplantation. 2004;78:434–441.
Mengel M, Reeve J, Bunnag S, et al. Molecular correlates of scarring in kidney transplants: the emergence of mast cell transcripts. Am J Transplant. 2009;9:169–178.
Mengel M, Reeve J, Bunnag S, et al. Scoring total inflammation is superior to the current Banff inflammation score in predicting outcome and the degree of molecular disturbance in renal allografts. Am J Transplant. 2009;9:1859–1867.
Sellarés J, de Freitas DG, Mengel M, et al. Inflammation lesions in kidney transplant biopsies: association with survival is due to the underlying diseases. Am J Transplant. 2011;11:489–499.
Mannon RB, Matas AJ, Grande J, et al. Inflammation in areas of tubular atrophy in kidney allograft biopsies: a potent predictor of allograft failure. Am J Transplant. 2010;10:2066–2073.
Nankivell BJ, Shingde M, Keung KL, et al. The causes, significance and consequences of inflammatory fibrosis in kidney transplantation: the Banff i-IFTA lesion. Am J Transplant. 2018;18:364–376.
Lefaucheur C, Gosset C, Rabant M, et al. T cell–mediated rejection is a major determinant of inflammation in scarred areas in kidney allografts. Am J Transplant. 2018;18:377–390.
Bouatou Y, Viglietti D, Pievani D, et al. Response to treatment and long-term outcomes in kidney transplant recipients with acute T cell–mediated rejection. Am J Transplant. 2019;19:1972–1988.
Nakagawa K, Tsuchimoto A, Ueki K, et al. Significance of revised criteria for chronic active T cell–mediated rejection in the 2017 Banff classification: surveillance by 1-year protocol biopsies for kidney transplantation. Am J Transplant. 2021;21:174–185.
Lamarche C, Cote J, Senecal L, et al. Efficacy of acute cellular rejection treatment according to Banff score in kidney transplant recipients: a systematic review. Transplant Direct. 2016;2:e115.
Ho J, Okoli GN, Rabbani R, et al. Effectiveness of T cell–mediated rejection therapy: a systematic review and meta-analysis. Am J Transplant. 2022;22:772–785.
Rampersad C, Balshaw R, Gibson IW, et al. The negative impact of T cell–mediated rejection on renal allograft survival in the modern era. Am J Transplant. 2022;22:761–771.
Aziz F, Parajuli S, Garg N, et al. How should acute T-cell mediated rejection of kidney transplants be treated: Importance of follow-up biopsy. Transplant Direct. 2022;8:e1305.
Chandran S, Leung J, Hu C, et al. Interleukin-6 blockade with tocilizumab increases Tregs and reduces T effector cytokines in renal graft inflammation: a randomized controlled trial. Am J Transplant. 2021;21:2543–2554.
Gaber A, First M, Tesi R, et al. Results of the double-blind, randomized, multicenter, phase iii clinical trial of thymoglobulin versus atgam in the treatment of acute graft rejection episodes after renal transplantation. Transplantation. 1998;66:29–37.
Kasiske BL, Zeier MG, Chapman JR, et al. KDIGO clinical practice guideline for the care of kidney transplant recipients: a summary. Kidney Int. 2010;77:299–311.
Loupy A, Vernerey D, Tinel C, et al. Subclinical rejection phenotypes at 1 year post-transplant and outcome of kidney allografts. J Am Soc Nephrol. 2015;26:1721.
Rush D, Nickerson P, Gough J, et al. Beneficial effects of treatment of early subclinical rejection: a randomized study. J Am Soc Nephrol. 1998;9:2129–2134.
Kurtkoti J, Sakhuja V, Sud K, et al. The utility of 1- and 3-month protocol biopsies on renal allograft function: a randomized controlled study. Am J Transplant. 2008;8:317–323.
Rush D, Arlen D, Boucher A, et al. Lack of benefit of early protocol biopsies in renal transplant patients receiving TAC and MMF: a randomized study. Am J Transplant. 2007;7:2538–2545.
Leblanc J, Subrt P, Paré M, et al. Practice patterns in the treatment and monitoring of acute T cell-mediated kidney graft rejection in Canada. Can J Kidney Health Dis. 2018;5:2054358117753616.
Park S, Guo K, Heilman RL, et al. Combining blood gene expression and cellfree DNA to diagnose subclinical rejection in kidney transplant recipients. Clin J Am Soc Nephrol. 2021;16:1539–1551.
Bu L, Gupta G, Pai A, et al. Clinical outcomes from the Assessing Donor-derived Cell-free DNA Monitoring Insights of Kidney Allografts with Longitudinal Surveillance (ADMIRAL) study. Kidney Int. 2022;101:793–803.
Gupta G, Moinuddin I, Kamal L, et al. Correlation of donor-derived cell-free DNA with histology and molecular diagnoses of kidney transplant biopsies. Transplantation. 2022;106:1061–1070.
Huang E, Haas M, MD P, et al. An assessment of the value of donor-derived cell-free DNA surveillance in patients with preserved kidney allograft function. Transplantation. 2022. doi:10.1097/TP.0000000000004267. (PMID: 10.1097/TP.0000000000004267)
Kung VL, Sandhu R, Haas M, et al. Chronic active T cell-mediated rejection is variably responsive to immunosuppressive therapy. Kidney Int. 2021;100:391–400.
Noguchi H, Matsukuma Y, Nakagawa K, et al. Treatment of chronic active T cell-mediated rejection after kidney transplantation: a retrospective cohort study of 37 transplants. Nephrology. 2022;27:632–638.
المشرفين على المادة: 0 (Isoantibodies)
تواريخ الأحداث: Date Created: 20221230 Date Completed: 20230427 Latest Revision: 20230523
رمز التحديث: 20240628
DOI: 10.1097/TP.0000000000004438
PMID: 36584369
قاعدة البيانات: MEDLINE
الوصف
تدمد:1534-6080
DOI:10.1097/TP.0000000000004438