Effect of Optimized Immunosuppression (Including Rituximab) on Anti-Donor Alloresponses in Patients With Chronically Rejecting Renal Allografts

Kin Yee Shiu, Dominic Stringer, Laura McLaughlin, Olivia Shaw, Paul Brookes, Hannah Burton, Hannah Wilkinson, Harriet Douthwaite, Tjir-Li Tsui, Adam Mclean, Rachel Hilton, Sian Griffin, Colin Geddes, Simon Ball, Richard Baker, Candice Roufosse, Catherine Horsfield, Anthony Dorling, Kin Yee Shiu, Dominic Stringer, Laura McLaughlin, Olivia Shaw, Paul Brookes, Hannah Burton, Hannah Wilkinson, Harriet Douthwaite, Tjir-Li Tsui, Adam Mclean, Rachel Hilton, Sian Griffin, Colin Geddes, Simon Ball, Richard Baker, Candice Roufosse, Catherine Horsfield, Anthony Dorling

Abstract

RituxiCAN-C4 combined an open-labeled randomized controlled trial (RCT) in 7 UK centers to assess whether rituximab could stabilize kidney function in patients with chronic rejection, with an exploratory analysis of how B cell-depletion influenced T cell anti-donor responses relative to outcome. Between January 2007 and March 2015, 59 recruits were enrolled after screening, 23 of whom consented to the embedded RCT. Recruitment was halted when in a pre-specified per protocol interim analysis, the RCT was discovered to be significantly underpowered. This report therefore focuses on the exploratory analysis, in which we confirmed that when B cells promoted CD4+ anti-donor IFNγ production assessed by ELISPOT, this associated with inferior clinical outcome; these patterns were inhibited by optimized immunosuppression but not rituximab. B cell suppression of IFNγ production, which associated with number of transitional B cells and correlated with slower declines in kidney function was abolished by rituximab, which depleted transitional B cells for prolonged periods. We conclude that in this patient population, optimized immunosuppression but not rituximab promotes anti-donor alloresponses associated with favorable outcomes. Clinical Trial Registration: Registered with EudraCT (2006-002330-38) and www.ClinicalTrials.gov, identifier: NCT00476164.

Keywords: B lymphocytes; chronic rejection in renal transplant; donor specific antibody (DSA); kidney transplantation; rituximab.

Copyright © 2020 Shiu, Stringer, McLaughlin, Shaw, Brookes, Burton, Wilkinson, Douthwaite, Tsui, Mclean, Hilton, Griffin, Geddes, Ball, Baker, Roufosse, Horsfield and Dorling.

Figures

Figure 1
Figure 1
Consort diagram for RituxiCAN-C4 trial. *Indicates 47 patients included in the exploratory analysis. †According to pre-specified second interim per protocol analysis.
Figure 2
Figure 2
Response to optimized immunosuppression. Exploratory analysis comparing those who responded favorably to optimized IS with those who did not. Graphs are box and whisker plots showing median with interquartile range (IQR) with whiskers showing upper and lower limits and outliers indicated as single data points. Means are represented with “x.” Time points: 0, enrolment sample; EP-1, End phase 1; 0–36, months post enrolment. White bars (n = 15); patients who responded well to optimized IS. Gray bars (n = 31 pre-enrolment. n = 32 post (one recruit did not have sufficient pre-enrolment creatinines); patients who failed to respond to optimized IS. (A) Urine PCR, (B) Tacrolimus trough levels, (C,D) Systolic, (C) and diastolic, (D) blood pressure (BP), (E,F) ΔeGFR normalized to enrolment ΔeGFR of 0. (G) Changes in Median Fluorescence Intensity (MFI) of cumulative DSA with time (NB includes values where DSA = 0). P-values by Mann Whitney U-test.
Figure 3
Figure 3
Changes in B cells with rituximab—data from RCT per protocol groups. Graphs are box and whisker plots showing median with interquartile range (IQR) with whiskers showing upper and lower limits and outliers indicated as single data points. Means are represented with “x.” Time points: 0= enrolment sample. EP-1, End phase 1; EP-2, End phase 2; 0–36, months post enrolment. Rituximab administered between EP-1 and EP-2. The gating strategy is described in detail in methods. “N” refers to the number of samples at each time point. (A–F) Changes in B cells in RCT. (A) Absolute numbers of B cells per uL of serum. (B–F) Flow cytometric analysis of the proportion of B cell subpopulations against time. (B) CD27-negative B cells as proportion of total CD19+ cells. (C) CD27+ B cells as proportion of CD19+ cells. (D) CD38loCD24lo cells as proportion of CD27- cells (naïve B cells). (E) CD38++CD24++cells as proportion of CD27- cells (Transitional T1 cells). Median absolute number of T1 per μL is shown beneath each column. (F) CD38+CD324+ cells as proportion of CD27- cells (Transitional T2 cells). P-values by Mann Whitney U-test.
Figure 4
Figure 4
ELISPOT patterns. (A–E) Illustrates the 3 basic patterns of anti-donor IFNγ production, displayed as the spot count (corrected for flow cytometric assessment of CD4+ cell proportions) present under 4 different conditions: CD8- (CD8-depleted PBMC); CD19- (CD8- & CD19-depleted PBMC), both performed in presence or absence of CD25+ cells. Samples showing anti-donor responsiveness from 51 recruits, including from those not in the exploratory analysis, are represented. (A) Pattern 1: Unregulated B cell-dependent pattern. Showing spot counts that reduce (>20%) on depletion of CD19+ cells in presence of CD25+ cells (± in absence of CD25+ cells). N = 16 samples. (B,C) Pattern 2: B cell-dependent anti-donor patterns with evidence of regulation. (B) CD25+ regulated B-dependent responses: B cell-dependent anti-donor responses only detectable in absence of CD25+ cells. N = 14 samples. (C) CD19+ regulated B-dependent responses. B cell-dependent anti-donor responses in presence of CD25+ cells, but when CD25+ cells absent, depletion of CD19+ cells increases spot count (>20%), indicating evidence of regulation by B cells. N = 2 samples. (D,E) Pattern 3: Regulated anti-donor responses without evidence of B cell-dependency. (D) CD19+ regulated responses; In presence of CD25+ cells, spot counts increase (>20%) when CD19+ cells are depleted. N = 11 samples. (E) CD25+ and CD19+ regulated. In absence of CD25+ cells, depletion of CD19+ cells increases spot counts (>20%). In presence of CD25+ cells, anti-donor responses are undetectable. N = 7 samples.
Figure 5
Figure 5
Associations with ELISPOT patterns. Graphs show box plots of median with IQR with whiskers showing upper and lower limits and outliers indicated as single data points. Means are represented with “x.” (A) Association between proportion of CD4+CD25+CD39hi T cells (Tregs) and ELISPOT patterns characterized by spot count suppression when CD25+ cells present. (B,C) Association between proportion of CD19+ cells having the phenotype of transitional T1 cells (CD27-CD38++CD24++) (B) or transitional T2 cells (CD27-CD38+CD24+) (C) and ELISPOT patterns showing evidence of increasing spot counts after depletion of CD19+ cells. (D,E) ΔeGFR, normalized to enrolment eGFR of 0 (D) and urine PCR (E) in patients with at least two samples at end-phase 2 or beyond (n = 27). White bars are patients who had either donor non-responsiveness or ELISPOT patterns with evidence of regulated anti-donor responses in their post-optimization samples (n = 21). Gray bars are those with at least one post-end-phase 2 sample showing evidence of unregulated B cell dependent anti-donor responses (n = 6). Time points: 0, enrolment sample; EP-1, End phase 1; EP-2, End phase 2; 0–36, months post enrolment. P-values by Mann Whitney U-test.

References

    1. Wang JH, Skeans MA, Israni AK. Current status of kidney transplant outcomes: dying to survive. Adv Chronic Kidney Dis. (2016) 23:281–6. 10.1053/j.ackd.2016.07.001
    1. Lamb KE, Lodhi S, Meier-Kriesche HU. Long-term renal allograft survival in the United States: a critical reappraisal. Am J Transplant. (2011) 11:450–62. 10.1111/j.1600-6143.2010.03283.x
    1. Ravanan R, Udayaraj U, Bakran A, Steenkamp R, Williams AJ, Ansell D. Measures of care in adult renal transplant recipients in the United Kingdom (chapter 11). Nephrol Dial Transplant. (2007) 22(Suppl. 7):vii:138–54. 10.1093/ndt/gfm334
    1. Gaston RS, Cecka JM, Kasiske BL, Fieberg AM, Leduc R, Cosio FC, et al. . Evidence for antibody-mediated injury as a major determinant of late kidney allograft failure. Transplantation. (2010) 90:68–74. 10.1097/TP.0b013e3181e065de
    1. Haas M, Sis B, Racusen LC, Solez K, Glotz D, Colvin RB, et al. . Banff 2013 meeting report: inclusion of c4d-negative antibody-mediated rejection and antibody-associated arterial lesions. Am J Transplant. (2014) 14:272–83. 10.1111/ajt.12590
    1. Worthington JE, Martin S, Al-Husseini DM, Dyer PA, Johnson RW. Posttransplantation production of donor HLA-specific antibodies as a predictor of renal transplant outcome. Transplantation. (2003) 75:1034–40. 10.1097/01.TP.0000055833.65192.3B
    1. Mizutani K, Terasaki P, Rosen A, Esquenazi V, Miller J, Shih RN, et al. . Serial ten-year follow-up of HLA and MICA antibody production prior to kidney graft failure. Am J Transplant. (2005) 5:2265–72. 10.1111/j.1600-6143.2005.01016.x
    1. Lee PC, Zhu L, Terasaki PI, Everly MJ. HLA-specific antibodies developed in the first year posttransplant are predictive of chronic rejection and renal graft loss. Transplantation. (2009) 88:568–74. 10.1097/TP.0b013e3181b11b72
    1. Dorling A. Transplant accommodation–are the lessons learned from xenotransplantation pertinent for clinical allotransplantation? Am J Transplant. (2012) 12:545–53. 10.1111/j.1600-6143.2011.03821.x
    1. Lefaucheur C, Viglietti D, Bentlejewski C, Duong van Huyen JP, Vernerey D, Aubert O, et al. . IgG donor-specific anti-human HLA antibody subclasses and kidney allograft antibody-mediated injury. J Am Soc Nephrol. (2016) 27:293–304. 10.1681/ASN.2014111120
    1. Loupy A, Lefaucheur C, Vernerey D, Prugger C, Duong van Huyen JP, Mooney N, et al. . Complement-binding anti-HLA antibodies and kidney-allograft survival. N Engl J Med. (2013) 369:1215–26. 10.1056/NEJMoa1302506
    1. Chong AS, Rothstein DM, Safa K, Riella LV. Outstanding questions in transplantation: B cells, alloantibodies, and humoral rejection. Am J Transplant. (2019) 19:2155–63. 10.1111/ajt.15323
    1. Shiu KY, McLaughlin L, Rebollo-Mesa I, Zhao J, Semik V, Cook HT, et al. . B-lymphocytes support and regulate indirect T-cell alloreactivity in individual patients with chronic antibody-mediated rejection. Kidney Int. (2015) 88:560–8. 10.1038/ki.2015.100
    1. Shiu KY, McLaughlin L, Rebollo-Mesa I, Zhao J, Burton H, Douthwaite H, et al. . Graft dysfunction in chronic antibody-mediated rejection correlates with B-cell-dependent indirect antidonor alloresponses and autocrine regulation of interferon-gamma production by Th1 cells. Kidney Int. (2017) 91:477–92. 10.1016/j.kint.2016.10.009
    1. Smith MR. Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance. Oncogene. (2003) 22:7359–68. 10.1038/sj.onc.1206939
    1. Barnett AN, Hadjianastassiou VG, Mamode N. Rituximab in renal transplantation. Transpl Int. (2013) 26:563–75. 10.1111/tri.12072
    1. Billing H, Rieger S, Ovens J, Susal C, Melk A, Waldherr R, et al. . Successful treatment of chronic antibody-mediated rejection with IVIG and rituximab in pediatric renal transplant recipients. Transplantation. (2008) 86:1214–21. 10.1097/TP.0b013e3181880b35
    1. Fehr T, Rusi B, Fischer A, Hopfer H, Wuthrich 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. Rostaing L, Guilbeau-Frugier C, Fort M, Mekhlati L, Kamar N. Treatment of symptomatic transplant glomerulopathy with rituximab. Transpl Int. (2009) 22:906–13. 10.1111/j.1432-2277.2009.00896.x
    1. Kamar N, Milioto O, Puissant-Lubrano B, Esposito L, Pierre MC, Mohamed AO, et al. . Incidence and predictive factors for infectious disease after rituximab therapy in kidney-transplant patients. Am J Transplant. (2010) 10:89–98. 10.1111/j.1600-6143.2009.02785.x
    1. Genberg H, Hansson A, Wernerson A, Wennberg L, Tyden G. Pharmacodynamics of rituximab in kidney allotransplantation. Am J Transplant. (2006) 6:2418–28. 10.1111/j.1600-6143.2006.01497.x
    1. Kamburova EG, Koenen HJ, van den Hoogen MW, Baas MC, Joosten I, Hilbrands LB. Longitudinal analysis of T and B cell phenotype and function in renal transplant recipients with or without rituximab induction therapy. PLoS ONE. (2014) 9:e112658 10.1371/journal.pone.0112658
    1. Zarkhin V, Lovelace PA, Li L, Hsieh SC, Sarwal MM. Phenotypic evaluation of B-cell subsets after rituximab for treatment of acute renal allograft rejection in pediatric recipients. Transplantation. (2011) 91:1010–8. 10.1097/TP.0b013e318213df29
    1. Sidner RA, Book BK, Agarwal A, Bearden CM, Vieira CA, Pescovitz MD. In vivo human B-cell subset recovery after in vivo depletion with rituximab, anti-human CD20 monoclonal antibody. Hum Antibodies. (2004) 13:55–62. 10.3233/HAB-2004-13301
    1. Ikemiyagi M, Hirai T, Ishii R, Miyairi S, Okumi M, Tanabe K. Transitional B cells predominantly reconstituted after a desensitization therapy using rituximab before kidney transplantation. Ther Apher Dial. (2017) 21:139–49. 10.1111/1744-9987.12508
    1. Sentis A, Diekmann F, Llobell A, de Moner N, Espinosa G, Yague J, et al. . Kinetic analysis of changes in T- and B-lymphocytes after anti-CD20 treatment in renal pathology. Immunobiology. (2017) 222:620–30. 10.1016/j.imbio.2016.11.011
    1. Blair PA, Norena LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, et al. . CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity. (2010) 32:129–40. 10.1016/j.immuni.2009.11.009
    1. Cherukuri A, Salama AD, Carter CR, Landsittel D, Arumugakani G, Clark B, et al. . Reduced human transitional B cell T1/T2 ratio is associated with subsequent deterioration in renal allograft function. Kidney Int. (2017) 91:183–95. 10.1016/j.kint.2016.08.028
    1. Dudley C, Pohanka E, Riad H, Dedochova J, Wijngaard P, Sutter C, et al. . Mycophenolate mofetil substitution for cyclosporine a in renal transplant recipients with chronic progressive allograft dysfunction: the “creeping creatinine” study. Transplantation. (2005) 79:466–75. 10.1097/01.TP.0000151632.21551.00
    1. Marx A, Backes C, Meese E, Lenhof HP, Keller A. EDISON-WMW: exact dynamic programing solution of the wilcoxon-mann-whitney test. Genomics Proteomics Bioinformatics. (2016) 14:55–61. 10.1016/j.gpb.2015.11.004
    1. Willicombe M, Brookes P, Sergeant R, Santos-Nunez E, Steggar C, Galliford J, et al. . De novo DQ donor-specific antibodies are associated with a significant risk of antibody-mediated rejection and transplant glomerulopathy. Transplantation. (2012) 94:172–7. 10.1097/TP.0b013e3182543950
    1. Baker RJ, Hernandez-Fuentes MP, Brookes PA, Chaudhry AN, Cook HT, Lechler RI. Loss of direct and maintenance of indirect alloresponses in renal allograft recipients: implications for the pathogenesis of chronic allograft nephropathy. J Immunol. (2001) 167:7199–206. 10.4049/jimmunol.167.12.7199
    1. Sagoo P, Perucha E, Sawitzki B, Tomiuk S, Stephens DA, Miqueu P, et al. . Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest. (2010) 120:1848–61. 10.1172/JCI39922
    1. Opelz G. Non-HLA transplantation immunity revealed by lymphocytotoxic antibodies. Lancet. (2005) 365:1570–6. 10.1016/S0140-6736(05)66458-6
    1. El-Zoghby ZM, Stegall MD, Lager DJ, Kremers WK, Amer H, Gloor JM, et al. . Identifying specific causes of kidney allograft loss. Am J Transplant. (2009) 9:527–35. 10.1111/j.1600-6143.2008.02519.x
    1. Macklin PS, Morris PJ, Knight SR. A systematic review of the use of rituximab for the treatment of antibody-mediated renal transplant rejection. Transplant Rev. (2017) 31:87–95. 10.1016/j.trre.2017.01.002
    1. Pineiro GJ, De Sousa-Amorim E, Sole M, Rios J, Lozano M, Cofan F, et al. . Rituximab, plasma exchange and immunoglobulins: an ineffective treatment for chronic active antibody-mediated rejection. BMC Nephrol. (2018) 19:261. 10.1186/s12882-018-1057-4
    1. Mella A, Gallo E, Messina M, Caorsi C, Amoroso A, Gontero P, et al. . Treatment with plasmapheresis, immunoglobulins and rituximab for chronic-active antibody-mediated rejection in kidney transplantation: Clinical, immunological and pathological results. World J Transplant. (2018) 8:178–87. 10.5500/wjt.v8.i5.178
    1. Moreso F, Crespo M, Ruiz JC, Torres A, Gutierrez-Dalmau A, Osuna A, et al. . Treatment of chronic antibody mediated rejection with intravenous immunoglobulins and rituximab: a multicenter, prospective, randomized, double-blind clinical trial. Am J Transplant. (2018) 18:927–35. 10.1111/ajt.14520
    1. Theruvath TP, Saidman SL, Mauiyyedi S, Delmonico FL, Williams WW, Tolkoff-Rubin N, et al. . Control of antidonor antibody production with tacrolimus and mycophenolate mofetil in renal allograft recipients with chronic rejection. Transplantation. (2001) 72:77–83. 10.1097/00007890-200107150-00016
    1. Zarkhin V, Li L, Kambham N, Sigdel T, Salvatierra O, Sarwal MM. A randomized, prospective trial of rituximab for acute rejection in pediatric renal transplantation. Am J Transplant. (2008) 8:2607–17. 10.1111/j.1600-6143.2008.02411.x
    1. Gokmen MR, Lombardi G, Lechler RI. The importance of the indirect pathway of allorecognition in clinical transplantation. Curr Opin Immunol. (2008) 20:568–74. 10.1016/j.coi.2008.06.009
    1. Gu J, Ni X, Pan X, Lu H, Lu Y, Zhao J, et al. Human CD39(hi) regulatory T cells present stronger stability and function under inflammatory conditions. Cell Mol Immunol. (2017) 14:521–8. 10.1038/cmi.2016.30
    1. Clatworthy MR, Watson CJ, Plotnek G, Bardsley V, Chaudhry AN, Bradley JA, et al. . B-cell-depleting induction therapy and acute cellular rejection. N Engl J Med. (2009) 360:2683–5. 10.1056/NEJMc0808481
    1. Parajuli S, Redfield RR, Garg N, Aziz F, Mohamed M, Astor BC, et al. . Clinical significance of microvascular inflammation in the absence of Anti-HLA DSA in kidney transplantation. Transplantation. (2018) 103:1468–76. 10.1097/TP.0000000000002487
    1. Debiais C, Goujon JM, Quellard N, Fernandez B, Milin S, Jollet I, et al. Glomerular C4d staining and chronic transplant glomerulopathy. Am J Trans. (2011) 11:296.
    1. Govil A, Everly M, Arend L, Everly J, Brailey P, Mogilishetty G, et al. Glomerular C4D deposition and de novo donor specific antibodies are associated with poor prognosis in transplant glomerulopathy. Am J Trans. (2009) 9:589.
    1. Sijpkens YW, Joosten SA, Wong MC, Dekker FW, Benediktsson H, Bajema IM, et al. . Immunologic risk factors and glomerular C4d deposits in chronic transplant glomerulopathy. Kidney Int. (2004) 65:2409–18. 10.1111/j.1523-1755.2004.00662.x
    1. Galliford J, Cook HT, Brookes PA, Chan K, Taube D, Dorling A. Reversal of refractory c4d chronic allograft nephropathy with rituximab. Am J Trans. (2007) 7(Suppl. 2):526.

Source: PubMed

3
購読する