Histologic and Molecular Patterns in Responders and Non-responders With Chronic-Active Antibody-Mediated Rejection in Kidney Transplants

Onur Sazpinar, Ariana Gaspert, Daniel Sidler, Markus Rechsteiner, Thomas F Mueller, Onur Sazpinar, Ariana Gaspert, Daniel Sidler, Markus Rechsteiner, Thomas F Mueller

Abstract

Introduction: There is no proven therapy for chronic-active antibody-mediated rejection (caABMR), the major cause of late kidney allograft failure. Histological and molecular patterns associated with possible therapy responsiveness are not known.

Methods: Based on rigorous selection criteria this single center, retrospective study identified 16 out of 1027 consecutive kidney transplant biopsies taken between 2008 and 2016 with pure, unquestionable caABMR, without other pathologic features. The change in estimated GFR pre- and post-biopsy/treatment were utilized to differentiate subjects into responders and non-responders. Gene sets reflecting active immune processes of caABMR were defined a priori, including endothelial, inflammatory, cellular, interferon gamma (IFNg) and calcineurin inhibitor (CNI) related-genes based on the literature. Transcript measurements were performed in RNA extracted from stored, formalin-fixed, paraffin-embedded (FFPE) samples using NanoString technology. Histology and gene expression patterns of responders and non-responders were compared.

Results: A reductionist approach applying very tight criteria to identify caABMR and treatment response excluded the vast majority of clinical ABMR cases. Only 16 out of 139 cases with a written diagnosis of chronic rejection fulfilled the caABMR criteria. Histological associations with therapy response included a lower peritubular capillaritis score (p = 0.028) along with less glomerulitis. In contrast, no single gene discriminated responders from non-responders. Activated genes associated with NK cells and endothelial cells suggested lack of treatment response.

Conclusion: In caABMR active microvascular injury, in particular peritubular capillaritis, differentiates treatment responders from non-responders. Transcriptome changes in NK cell and endothelial cell associated genes may further help to identify treatment response. Future prospective studies will be needed which include more subjects, who receive standardized treatment protocols to identify biomarkers for treatment response.

Clinical trial registration: [ClinicalTrials.gov], identifier [NCT03430414].

Keywords: Banff classification; chronic-active ABMR; eGFR slope; kidney transplantation; therapy response; transcriptome.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Sazpinar, Gaspert, Sidler, Rechsteiner and Mueller.

Figures

FIGURE 1
FIGURE 1
Algorithm of the biopsy selection process. cABMR, chronic antibody mediated rejection; aABMR, active antibody mediated rejection; IF/TA, interstitial fibrosis and tubular atrophy; ICT, information and communication technology; caABMR, chronic-active antibody mediated rejection fulfilling Banff 2017 criteria; ptc, peritubular capillaritis; g, glomerulitis; cg, transplant glomerulopathy; C4d, complement split product.
FIGURE 2
FIGURE 2
Kidney function before and after biopsy in treatment responders versus non-responders. (A) eGFR slopes in 10 therapy responders, (B) in 6 non-responders. Every line represents one patient. The linearized slopes were calculated by linear regression of raw data before and after biopsy and plotting of the two curves ypre–biopsy = mpre–biopsy *x + qpre–biopsy and y = mpost–biopsy*x + qpost–biopsy where qpre–biopsy = qpost–biopsy = measured eGFR at biopsy day.
FIGURE 3
FIGURE 3
Overview of Banff scores of responders versus non-responders. t, tubulitis; i, interstitial inflammation; ti, total cortical inflammation; ptc, peritubular capillaritis; v, vasculitis; cv, arterial fibrous intimal thickening; g, glomerulitis; cg, transplant glomerulopathy; mm, mesangial matrix expansion; ci, interstitial fibrosis; ct, tubular atrophy; ah, arteriolar hyalinosis; aah, hyaline arteriolar thickening; C4d, complement split product; IFTA, interstitial fibrosis–tubular atrophy.
FIGURE 4
FIGURE 4
Clustering of histopathology in the caABMR biopsies. (A) Principal component analysis indicating distribution and correlation between Banff scores. (B) Unsupervised hierarchical clustering of the Banff characteristics based on the samples and (C) of the biopsies based on 8 Banff features. Due to high Euclidian distances 3 samples did not include in the analysis.
FIGURE 5
FIGURE 5
Clustering of transcriptomes in the caABMR biopsies. (A) Unsupervised hierarchical clustering of the 44 gene transcripts. The dendrogram shows the clustering of the 44 genes according to similarities and dissimilarities. (B) Dendrogram of distribution of responders and non-responders based on gene expression.
FIGURE 6
FIGURE 6
Heatmap analysis of gene expression. Columns represent the 16 different samples, rows the 44 analyzed genes. High gene expression is shown in green, low gene expression in red. Similarity metric was calculated with uncentered Pearson correlation for patients/biopsies and genes; average linkage was the used linkage method.

References

    1. Loupy A, Hill GS, Jordan SC. The impact of donor-specific anti-HLA antibodies on late kidney allograft failure. Nat Rev Nephrol. (2012) 8:348–57. 10.1038/nrneph.2012.81
    1. Suri DL, Tomlanovich SJ, Olson JL, Meyer TW. Transplant glomerulopathy as a cause of late graft loss. Am J Kidney Dis. (2000) 35:674–80. 10.1016/s0272-6386(00)70015-1
    1. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant. (2009) 9(Suppl. 3): S1–155.
    1. Madill-Thomsen K, Perkowska-Ptasińska A, Böhmig GA, Eskandary F, Einecke G, Gupta G, et al. Discrepancy analysis comparing molecular and histology diagnoses in kidney transplant biopsies. Am J Transplant. (2020) 20:1341–50. 10.1111/ajt.15752
    1. Böhmig GA, Eskandary F, Doberer K, Halloran PF. The therapeutic challenge of late antibody-mediated kidney allograft rejection. Transpl Int. (2019) 32:775–88. 10.1111/tri.13436
    1. Stegall MD, Gaston RS, Cosio FG, Matas A. Through a glass darkly: seeking clarity in preventing late kidney transplant failure. J Am Soc Nephrol. (2015) 26:20–9. 10.1681/ASN.2014040378
    1. Birnbaum LM, Lipman M, Paraskevas S, Chaudhury P, Tchervenkov J, Baran D, et al. Management of chronic allograft nephropathy: a systematic review. Clin J Am Soc Nephrol. (2009) 4:860–5.
    1. Roberts DM, Jiang SH, Chadban SJ. The treatment of acute antibody-mediated rejection in kidney transplant recipients-a systematic review. Transplantation. (2012) 94:775–83. 10.1097/TP.0b013e31825d1587
    1. Parajuli S, Joachim E, Alagusundaramoorthy S, Blazel J, Aziz F, Garg N, et al. Subclinical antibody-mediated rejection after kidney transplantation: treatment outcomes. Transplantation. (2019) 103:1722–9. 10.1097/TP.0000000000002566
    1. Perrottet N, Fernández-Ruiz M, Binet I, Dickenmann M, Dahdal S, Hadaya K, et al. Infectious complications and graft outcome following treatment of acute antibody-mediated rejection after kidney transplantation: a nationwide cohort study. PLoS One. (2021) 16:e0250829. 10.1371/journal.pone.0250829
    1. O’Connell PJ, Zhang W, Menon MC, Yi Z, Schröppel B, Gallon L, et al. Biopsy transcriptome expression profiling to identify kidney transplants at risk of chronic injury: a multicentre, prospective study. Lancet. (2016) 388:983–93. 10.1016/S0140-6736(16)30826-1
    1. Crespo E, Roedder S, Sigdel T, Hsieh SC, Luque S, Cruzado JM, et al. Molecular and functional noninvasive immune monitoring in the ESCAPE study for prediction of subclinical renal allograft rejection. Transplantation. (2017) 101:1400–9. 10.1097/TP.0000000000001287
    1. Sellarés J, Reeve J, Loupy A, Mengel M, Sis B, Skene A, et al. Molecular diagnosis of antibody-mediated rejection in human kidney transplants. Am J Transplant. (2013) 13:971–83. 10.1111/ajt.12150
    1. Mueller TF, Solez K, Mas V. Assessment of kidney organ quality and prediction of outcome at time of transplantation. Semin Immunopathol. (2011) 33:185–99. 10.1007/s00281-011-0248-x
    1. Kumar D, Raynaud M, Chang J, Reeve J, Yakubu I, Kamal L, et al. Impact of belatacept conversion on renal function, histology, and gene expression in kidney transplant patients with chronic active antibody-mediated rejection. Transplantation. (2021) 105:660–7. 10.1097/TP.0000000000003278
    1. Loupy A, Lefaucheur C, Vernerey D, Chang J, Hidalgo LG, Beuscart T, et al. Molecular microscope strategy to improve risk stratification in early antibody-mediated kidney allograft rejection. J Am Soc Nephrol. (2014) 25:2267–77. 10.1681/ASN.2013111149
    1. Venner JM, Hidalgo LG, Famulski KS, Chang J, Halloran PF. The molecular landscape of antibody-mediated kidney transplant rejection: evidence for NK involvement through CD16a Fc receptors. Am J Transplant. (2015) 15:1336–48. 10.1111/ajt.13115
    1. Afzali B, Chapman E, Racapé M, Adam B, Bruneval P, Gil F, et al. Molecular assessment of microcirculation injury in formalin-fixed human cardiac allograft biopsies with antibody-mediated rejection. Am J Transplant. (2017) 17:496–505. 10.1111/ajt.13956
    1. Haas M, Loupy A, Lefaucheur C, Roufosse C, Glotz D, Seron D, 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. 10.1111/ajt.14625
    1. Jordan SC, Quartel AW, Czer LS, Admon D, Chen G, Fishbein MC, et al. Posttransplant therapy using high-dose human immunoglobulin (intravenous gammaglobulin) to control acute humoral rejection in renal and cardiac allograft recipients and potential mechanism of action. Transplantation. (1998) 66:800–5. 10.1097/00007890-199809270-00017
    1. Luke PP, Scantlebury VP, Jordan ML, Vivas CA, Hakala TR, Jain A, et al. Reversal of steroid- and anti-lymphocyte antibody-resistant rejection using intravenous immunoglobulin (IVIG) in renal transplant recipients. Transplantation. (2001) 72:419–22. 10.1097/00007890-200108150-00010
    1. Bamoulid J, Staeck O, Halleck F, Dürr M, Paliege A, Lachmann N, et al. Advances in pharmacotherapy to treat kidney transplant rejection. Expert Opin Pharmacother. (2015) 16:1627–48. 10.1517/14656566.2015.1056734
    1. Casadei DH, del C Rial M, Opelz G, Golberg JC, Argento JA, Greco G, et al. A randomized and prospective study comparing treatment with high-dose intravenous immunoglobulin with monoclonal antibodies for rescue of kidney grafts with steroid-resistant rejection. Transplantation. (2001) 71:53–8. 10.1097/00007890-200101150-00009
    1. Fehr T, Rüsi B, Fischer A, Hopfer H, Wüthrich RP, Gaspert A. Rituximab and intravenous immunoglobulin treatment of chronic antibody-mediated kidney allograft rejection. Transplantation. (2009) 87:1837–41. 10.1097/TP.0b013e3181a6bac5
    1. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AFIII, Feldman HI, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. (2009) 150:604–12.
    1. Valenzuela NM, Reed EF. Antibody-mediated rejection across solid organ transplants: manifestations, mechanisms, and therapies. J Clin Invest. (2017) 127:2492–504. 10.1172/JCI90597
    1. Halloran PF, Venner JM, Madill-Thomsen KS, Einecke G, Parkes MD, Hidalgo LG, et al. Review: the transcripts associated with organ allograft rejection. Am J Transplant. (2018) 18:785–95. 10.1111/ajt.14600
    1. Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF. Endothelial gene expression in kidney transplants with alloantibody indicates antibody-mediated damage despite lack of C4d staining. Am J Transplant. (2009) 9:2312–23. 10.1111/j.1600-6143.2009.02761.x
    1. Zhang X, Reed EF. Effect of antibodies on endothelium. Am J Transplant. (2009) 9:2459–65. 10.1111/j.1600-6143.2009.02819.x
    1. Hirohashi T, Chase CM, Della Pelle P, Sebastian D, Alessandrini A, Madsen JC, et al. A novel pathway of chronic allograft rejection mediated by NK cells and alloantibody. Am J Transplant. (2012) 12:313–21. 10.1111/j.1600-6143.2011.03836.x
    1. Hidalgo LG, Sis B, Sellares J, Campbell PM, Mengel M, Einecke G, et al. NK cell transcripts and NK cells in kidney biopsies from patients with donor-specific antibodies: evidence for NK cell involvement in antibody-mediated rejection. Am J Transplant. (2010) 10:1812–22. 10.1111/j.1600-6143.2010.03201.x
    1. Parkes MD, Halloran PF, Hidalgo LG. Evidence for CD16a-mediated NK cell stimulation in antibody-mediated kidney transplant rejection. Transplantation. (2017) 101:e102–11. 10.1097/TP.0000000000001586
    1. Halloran PF, Afrouzian M, Ramassar V, Urmson J, Zhu LF, Helms LM, et al. Interferon-gamma acts directly on rejecting renal allografts to prevent graft necrosis. Am J Pathol. (2001) 158:215–26. 10.1016/s0002-9440(10)63960-0
    1. Tsang HF, Xue VW, Koh SP, Chiu YM, Ng LP, Wong SC. NanoString, a novel digital color-coded barcode technology: current and future applications in molecular diagnostics. Expert Rev Mol Diagn. (2017) 17:95–103. 10.1080/14737159.2017.1268533
    1. Schinstock CA, Mannon RB, Budde K, Chong AS, Haas M, Knechtle S, et al. Recommended treatment for antibody-mediated rejection after kidney transplantation: the 2019 expert consensus from the transplantion society working group. Transplantation. (2020) 104:911–22. 10.1097/TP.0000000000003095
    1. Mulley WR, Huang LL, Ramessur Chandran S, Longano A, Amos LAR, Polkinghorne KR, et al. Long-term graft survival in patients with chronic antibody-mediated rejection with persistent peritubular capillaritis treated with intravenous immunoglobulin and rituximab. Clin Transplant. (2017) 31:e13037. 10.1111/ctr.13037
    1. Burton SA, Amir N, Asbury A, Lange A, Hardinger KL. Treatment of antibody-mediated rejection in renal transplant patients: a clinical practice survey. Clin Transplant. (2015) 29:118–23. 10.1111/ctr.12491
    1. Sautenet B, Blancho G, Büchler M, Morelon E, Toupance O, Barrou B, et al. One-year results of the effects of rituximab on acute antibody-mediated rejection in renal transplantation: Ritux ERAH, a multicenter Double-blind randomized placebo-controlled trial. Transplantation. (2016) 100:391–9. 10.1097/TP.0000000000000958
    1. Parajuli S, Mandelbrot DA, Muth B, Mohamed M, Garg N, Aziz F, et al. Rituximab and monitoring strategies for late antibody-mediated rejection after kidney transplantation. Transplant Direct. (2017) 3:e227. 10.1097/TXD.0000000000000746
    1. Archdeacon P, Chan M, Neuland C, Velidedeoglu E, Meyer J, Tracy L, et al. Summary of FDA antibody-mediated rejection workshop. Am J Transplant. (2011) 11:896–906. 10.1111/j.1600-6143.2011.03525.x
    1. Loupy A, Lefaucheur C. Antibody-mediated rejection of solid-organ allografts. N Engl J Med. (2018) 379:1150–60. 10.1056/nejmra1802677
    1. McLaren AJ, Fuggle SV, Welsh KI, Gray DW, Morris PJ. Chronic allograft failure in human renal transplantation: a multivariate risk factor analysis. Ann Surg. (2000) 232:98–103. 10.1097/00000658-200007000-00014
    1. Reichel H, Zeier M, Ritz E. Proteinuria after renal transplantation: pathogenesis and management. Nephrol Dial Transplant. (2004) 19:301–5. 10.1093/ndt/gfh002
    1. Talreja H, Akbari A, White CA, Ramsay TO, Hiremath S, Knoll G. Predicting kidney transplantation outcomes using proteinuria ascertained from spot urine samples versus timed urine collections. Am J Kidney Dis. (2014) 64:962–8. 10.1053/j.ajkd.2014.07.027
    1. Einecke G, Sis B, Reeve J, Mengel M, Campbell PM, Hidalgo LG, et al. Antibody-mediated microcirculation injury is the major cause of late kidney transplant failure. Am J Transplant. (2009) 9:2520–31. 10.1111/j.1600-6143.2009.02799.x
    1. Cosio FG, Gloor JM, Sethi S, Stegall MD. Transplant glomerulopathy. Am J Transplant. (2008) 8:492–6.
    1. Aubert O, Higgins S, Bouatou Y, Yoo D, Raynaud M, Viglietti D, et al. Archetype analysis identifies distinct profiles in renal transplant recipients with transplant glomerulopathy associated with allograft survival. J Am Soc Nephrol. (2019) 30:625–39. 10.1681/ASN.2018070777
    1. Yazdani S, Callemeyn J, Gazut S, Lerut E, de Loor H, Wevers M, et al. Natural killer cell infiltration is discriminative for antibody-mediated rejection and predicts outcome after kidney transplantation. Kidney Int. (2019) 95:188–98. 10.1016/j.kint.2018.08.027
    1. Bontha SV, Maluf DG, Mueller TF, Mas VR. Systems biology in kidney transplantation: the application of multi-omics to a complex model. Am J Transplant. (2017) 17:11–21. 10.1111/ajt.13881
    1. Mas VR, Mueller TF, Archer KJ, Maluf DG. Identifying biomarkers as diagnostic tools in kidney transplantation. Expert Rev Mol Diagn. (2011) 11:183–96. 10.1586/erm.10.119
    1. GENE. Gene Integrates Information from a Wide Range of Species. A Record may Include Nomenclature, Reference Sequences (RefSeqs), Maps, Pathways, Variations, Phenotypes, and Links to Genome-, Phenotype-, and Locus-Specific Resources Worldwide. (2019) Available online at: (accessed July 15, 2019).
    1. Horuk R, Martin A, Hesselgesser J, Hadley T, Lu ZH, Wang ZX, et al. The Duffy antigen receptor for chemokines: structural analysis and expression in the brain. J Leukoc Biol. (1996) 59:29–38. 10.1002/jlb.59.1.29
    1. Segerer S, Regele H, MacK M, Kain R, Cartron JP, Colin Y, et al. The Duffy antigen receptor for chemokines is up-regulated during acute renal transplant rejection and crescentic glomerulonephritis. Kidney Int. (2000) 58:1546–56. 10.1046/j.1523-1755.2000.00316.x
    1. Adam BA, Smith RN, Rosales IA, Matsunami M, Afzali B, Oura T, et al. Chronic antibody-mediated rejection in nonhuman primate renal allografts: validation of human histological and molecular phenotypes. Am J Transplant. (2017) 17:2841–50. 10.1111/ajt.14327
    1. Hossain MN, Sakemura R, Fujii M, Ayusawa D. G-protein gamma subunit GNG11 strongly regulates cellular senescence. Biochem Biophys Res Commun. (2006) 351:645–50. 10.1016/j.bbrc.2006.10.112
    1. Nafar M, Kalantari S, Samavat S, Rezaei-Tavirani M, Rutishuser D, Zubarev RA. The novel diagnostic biomarkers for focal segmental glomerulosclerosis. Int J Nephrol. (2014) 2014:574261.
    1. Mannam VK, Lewis RE, Cruse JM. The fate of renal allografts hinges on responses of the microvascular endothelium. Exp Mol Pathol. (2013) 94:398–411. 10.1016/j.yexmp.2012.06.002
    1. Morgan JD, Lycett A, Horsburgh T, Nicholson ML, Veitch PS, Bell PR. The importance of E-selectin as a marker for renal transplant rejection. Transpl Immunol. (1994) 2:326–30. 10.1016/0966-3274(94)90010-8
    1. Suviolahti E, Ge S, Nast CC, Mirocha J, Karasyov A, White M, et al. Genes associated with antibody-dependent cell activation are overexpressed in renal biopsies from patients with antibody-mediated rejection. Transpl Immunol. (2015) 32:9–17. 10.1016/j.trim.2014.11.215
    1. Luyckx VA, Cairo LV, Compston CA, Phan WL, Mueller TF. Oncostatin M pathway plays a major role in the renal acute phase response. Am J Physiol Renal Physiol. (2009) 296:F875–83. 10.1152/ajprenal.90633.2008
    1. Yamada T. Serum amyloid A (SAA): a concise review of biology, assay methods and clinical usefulness. Clin Chem Lab Med. (1999) 37:381–8. 10.1515/CCLM.1999.063
    1. Müller TF, Trösch F, Ebel H, Grüssner RW, Feiber H, Göke B, et al. Pancreas-specific protein (PASP), serum amyloid A (SAA), and neopterin (NEOP) in the diagnosis of rejection after simultaneous pancreas and kidney transplantation. Transpl Int. (1997) 10:185–91. 10.1007/s001470050039
    1. Hocke G, Ebel H, Bittner K, Müller T, Kaffarnik H, Steinmetz A, et al. A rapid laser immunonephelometric assay for serum amyloid A (SAA) and its application to the diagnosis of kidney allograft rejection. Klin Wochenschr. (1989) 67:447–51. 10.1007/BF01725141
    1. Jordan SC, Choi J, Kim I, Wu G, Toyoda M, Shin B, et al. Interleukin-6, A cytokine critical to mediation of inflammation, autoimmunity and allograft rejection: therapeutic implications of IL-6 receptor blockade. Transplantation. (2017) 101:32–44. 10.1097/TP.0000000000001452
    1. Dean PG, Park WD, Cornell LD, Gloor JM, Stegall MD. Intragraft gene expression in positive crossmatch kidney allografts: ongoing inflammation mediates chronic antibody-mediated injury. Am J Transplant. (2012) 12:1551–63. 10.1111/j.1600-6143.2011.03964.x
    1. Zhu P, Xie L, Ding HS, Gong Q, Yang J, Yang L. High mobility group box 1 and kidney diseases (Review). Int J Mol Med. (2013) 31:763–8. 10.3892/ijmm.2013.1286
    1. Lv LL, Tang PM, Li CJ, You YK, Li J, Huang XR, et al. The pattern recognition receptor, mincle, is essential for maintaining the M1 macrophage phenotype in acute renal inflammation. Kidney Int. (2017) 91:587–602.
    1. Inoue T. M1 macrophage triggered by mincle leads to a deterioration of acute kidney injury. Kidney Int. (2017) 91:526–9. 10.1016/j.kint.2016.11.026
    1. Claus M, Herro R, Wolf D, Buscher K, Rudloff S, Huynh-Do U, et al. The TWEAK/Fn14 pathway is required for calcineurin inhibitor toxicity of the kidneys. Am J Transplant. (2018) 18:1636–45. 10.1111/ajt.14632

Source: PubMed

3
Suscribir