Distribution of de novo Donor-Specific Antibody Subclasses Quantified by Mass Spectrometry: High IgG3 Proportion Is Associated With Antibody-Mediated Rejection Occurrence and Severity

Vincent Pernin, Anais Beyze, Ilan Szwarc, Nicole Bec, Céline Salsac, Esther Perez-Garcia, Georges Mourad, Pierre Merville, Jonathan Visentin, Helene Perrochia, Christian Larroque, Lionel Couzi, Moglie Le Quintrec, Vincent Pernin, Anais Beyze, Ilan Szwarc, Nicole Bec, Céline Salsac, Esther Perez-Garcia, Georges Mourad, Pierre Merville, Jonathan Visentin, Helene Perrochia, Christian Larroque, Lionel Couzi, Moglie Le Quintrec

Abstract

Donor-specific antibodies (DSAs) are the main risk factor for antibody-mediated rejection (ABMR) and graft loss but could have variable pathogenicity according to their IgG subclass composition. Luminex-based test might lack sensitivity for the detection of IgG subclasses and this test does not allow quantifying the relative abundance of each IgG subclass. We investigated the precise repartition of each DSA subclass and their role in ABMR occurrence and severity, using an innovative mass spectrometry-based method. Between 2014 and 2018, we enrolled 69 patients who developed de novo DSA (n = 29 without ABMR, and n = 40 with ABMR) in two transplant centers. All IgG subclasses were detected in every samples tested: 62.7% were IgG1, 26.6% were IgG2, 6.6% were IgG3, and 4.2% were IgG4. The IgG3 proportion was significantly higher in the ABMR+ compared to the ABMR- group (8.4% vs. 5.6%, p = 0.003). The proportion of IgG1, IgG2, and IgG4 of DSA was similar between the two groups. Higher IgG3 level was associated with higher C4d deposition, higher microvascular inflammation scores, and glomerular filtration rate decline >25%. IgG3 proportion was not correlated with DSA MFI. Multivariate analysis showed that proteinuria and high level of IgG3 DSA were the only two factors independently associated with ABMR. In conclusion, de novo DSA are always composed of the four IgG subclasses, but in different proportions. High IgG3 proportion is associated with ABMR occurrence and severity and with poorer outcome, independently of DSA MFI.

Trial registration: ClinicalTrials.gov NCT04026087.

Keywords: IgG subclass; antibody-mediated rejection; de novo DSA; kidney transplantation; mass spectrometry.

Copyright © 2020 Pernin, Beyze, Szwarc, Bec, Salsac, Perez-Garcia, Mourad, Merville, Visentin, Perrochia, Larroque, Couzi and Le Quintrec.

Figures

Figure 1
Figure 1
Study flow chart.
Figure 2
Figure 2
Comparison of the IgG subclass distribution in total IgG and iDSA samples from all patients.
Figure 3
Figure 3
Comparison of the IgG subclass distribution in de novo iDSA isolated from DSA+/ABMR+ and DSA+/ABMR– patients. Proportion of IgG1 (A), IgG2 (B), IgG3 (C) and IgG4 (D) which composed iDSA in ABMR+ and ABMR− groups.
Figure 4
Figure 4
Percentage of IgG3 DSA is correlated with C4d deposition and microvascular inflammation scores.
Figure 5
Figure 5
Kaplan-Meyer curves for eGFR decline-free survival according to IgG3 level in all patients (A) and in DSA+ABMR+ group (B), and according to MFI level in all patients (C) and in DSA+ABMR+ group (D).

References

    1. Sellarés J, de Freitas DG, Mengel M, Reeve J, Einecke G, Sis B, et al. . Understanding the causes of kidney transplant failure: the dominant role of antibody-mediated rejection and nonadherence. Am J Transplant. (2012) 12:388–99. 10.1111/j.1600-6143.2011.03840.x
    1. Stegall MD, Chedid MF, Cornell LD. The role of complement in antibody-mediated rejection in kidney transplantation. Nat Rev Nephrol. (2012) 8:670–8. 10.1038/nrneph.2012.212
    1. Everly MJ, Rebellato LM, Haisch CE, Ozawa M, Parker K, Briley KP, et al. . Incidence and impact of de novo donor-specific alloantibody in primary renal allografts. Transplantation. (2013) 95:410–7. 10.1097/TP.0b013e31827d62e3
    1. Wiebe C, Gibson IW, Blydt-Hansen TD, Karpinski M, Ho J, Storsley LJ, et al. . Evolution and clinical pathologic correlations of de novo donor-specific HLA antibody post kidney transplant. Am J Transplant. (2012) 12:1157–67. 10.1111/j.1600-6143.2012.04013.x
    1. Lefaucheur C, Loupy A, Hill GS, Andrade J, Nochy D, Antoine C, et al. . Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol. (2010) 21:1398–406. 10.1681/ASN.2009101065
    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. Michaelsen TE, Garred P, Aase A. Human IgG subclass pattern of inducing complement-mediated cytolysis depends on antigen concentration and to a lesser extent on epitope patchiness, antibody affinity and complement concentration. Eur J Immunol. (1991) 21:11–16. 10.1002/eji.1830210103
    1. Bruhns P, Iannascoli B, England P, Mancardi DA, Fernandez N, Jorieux S, et al. . Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. Blood. (2009) 113:3716–25. 10.1182/blood-2008-09-179754
    1. Hönger G, Hopfer H, Arnold ML, Spriewald BM, Schaub S, Amico P. Pretransplant IgG subclasses of donor-specific human leukocyte antigen antibodies and development of antibody-mediated rejection. Transplantation. (2011) 92:41–7. 10.1097/TP.0b013e31821cdf0d
    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. Schnaidt M, Weinstock C, Jurisic M, Schmid-Horch B, Ender A, Wernet D. HLA antibody specification using single-antigen beads, a technical solution for the prozone effect. Transplantation. (2011) 92:510–5. 10.1097/TP.0b013e31822872dd
    1. Visentin J, Vigata M, Daburon S, Contin-Bordes C, Fremeaux-Bacchi V, Dromer C, et al. . Deciphering complement interference in anti-human leukocyte antigen antibody detection with flow beads assays. Transplantation. (2014) 98:625–31. 10.1097/TP.0000000000000315
    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. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of diet in renal disease study group. Ann Intern Med. (1999) 130:461–70. 10.7326/0003-4819-130-6-199903160-00002
    1. Dowle AA, Wilson J, Thomas JR. Comparing the diagnostic classification accuracy of iTRAQ, peak-area, spectral-counting, and emPAI methods for relative quantification in expression proteomics. J Proteome Res. (2016) 15:3550–62. 10.1021/acs.jproteome.6b00308
    1. Ladwig PM, Barnidge DR, Snyder MR, Katzmann JA, Murray DL. Quantification of serum IgG subclasses by use of subclass-specific tryptic peptides and liquid chromatography–tandem mass spectrometry. Clin Chem. (2014) 60:1080–8. 10.1373/clinchem.2014.222208
    1. Miyamoto S, Stroble CD, Taylor S, Hong Q, Lebrilla CB, Leiserowitz GS, et al. . Multiple reaction monitoring for the quantitation of serum protein glycosylation profiles: application to ovarian cancer. J Proteome Res. (2018) 17:222–33. 10.1021/acs.jproteome.7b00541
    1. Yuan W, Sanda M, Wu J, Koomen J, Goldman R. Quantitative analysis of immunoglobulin subclasses and subclass specific glycosylation by LC-MS-MRM in liver disease. J Proteomics. (2015) 116:24–33. 10.1016/j.jprot.2014.12.020
    1. Freitas MC, Rebellato LM, Ozawa M, Nguyen A, Sasaki N, Everly M, et al. . The role of immunoglobulin-G subclasses and C1q in de novo HLA-DQ donor-specific antibody kidney transplantation outcomes. Transplantation. (2013) 95:1113–9. 10.1097/TP.0b013e3182888db6
    1. Khovanova N, Daga S, Shaikhina T, Krishnan N, Jones J, Zehnder D, et al. . Subclass analysis of donor HLA-specific IgG in antibody-incompatible renal transplantation reveals a significant association of IgG4 with rejection and graft failure. Transpl Int. (2015) 28:1405–15. 10.1111/tri.12648
    1. Hamdani G, Goebel JW, Brailey P, Portwood EA, Hooper DK, Girnita AL. IgG3 anti-HLA donor-specific antibodies and graft function in pediatric kidney transplant recipients. Pediatr Transplant. (2018) 22:e13219. 10.1111/petr.13219
    1. Lowe D, Higgins R, Zehnder D, Briggs DC. Significant IgG subclass heterogeneity in HLA-specific antibodies: Implications for pathogenicity, prognosis, and the rejection response. Hum Immunol. (2013) 74:666–72. 10.1016/j.humimm.2013.01.008
    1. Siber GR, Schur PH, Aisenberg AC, Weitzman SA, Schiffman G. Correlation between serum IgG-2 concentrations and the antibody response to bacterial polysaccharide antigens. N Engl J Med. (1980) 303:178–82. 10.1056/NEJM198007243030402
    1. Smith TF. IgG subclasses. Adv Pediatr. (1992) 39:101–26.
    1. Aalberse RC, van der Gaag R, van Leeuwen J. Serologic aspects of IgG4 antibodies. Prolonged immunization results in an IgG4-restricted response. J Immunol. (1983) 130:722–6.
    1. Aversa G, Punnonen J, Carballido JM, Cocks BG, de Vries JE. CD40 ligand-CD40 interaction in Ig isotype switching in mature and immature human B cells. Semin Immunol. (1994) 6:295–301. 10.1006/smim.1994.1038
    1. Coffman RL, Lebman DA, Rothman P. Mechanism and regulation of immunoglobulin isotype switching. Adv Immunol. (1993) 54:229–70. 10.1016/S0065-2776(08)60536-2
    1. Avery DT, Bryant VL, Ma CS, de Waal Malefyt R, Tangye SG. IL-21-induced isotype switching to IgG and IgA by human naive B cells is differentially regulated by IL-4. J Immunol. (2008) 181:1767–79. 10.4049/jimmunol.181.3.1767
    1. Everly MJ, Rebellato LM, Haisch CE, Briley KP, Bolin P, Kendrick WT, et al. . Impact of IgM and IgG3 anti-HLA alloantibodies in primary renal allograft recipients. Transplantation. (2014) 97:494–501. 10.1097/01.TP.0000441362.11232.48
    1. Kaneku H, O'Leary JG, Taniguchi M, Susskind BM, Terasaki PI, Klintmalm GB. Donor-specific human leukocyte antigen antibodies of the immunoglobulin G3 subclass are associated with chronic rejection and graft loss after liver transplantation. Liver Transplant. (2012) 18:884–892. 10.1002/lt.23451
    1. O'Leary JG, Kaneku H, Banuelos N, Jennings LW, Klintmalm GB, Terasaki PI. Impact of IgG3 subclass and C1q-fixing donor-specific HLA alloantibodies on rejection and survival in liver transplantation. Am J Transplant. (2015) 15:1003–13. 10.1111/ajt.13153
    1. Schinstock CA, Cosio F, Cheungpasitporn W, Dadhania DM, Everly MJ, Samaniego-Picota MD, et al. . The value of protocol biopsies to identify patients with de novo donor-specific antibody at high risk for allograft loss. Am J Transplant. (2017) 17:1574–84. 10.1111/ajt.14161
    1. Kwon H, Kim YH, Choi JY, Shin S, Jung JH, Park SK, et al. . Impact of pretransplant donor-specific antibodies on kidney allograft recipients with negative flow cytometry cross-matches. Clin Transplant. (2018) 32:e13266. 10.1111/ctr.13266
    1. de Castro MCR, Barbosa EA, Souza RP, et al. . The kinetics of anti-hla antibodies in the first year after kidney transplantation: in whom and when should they be monitored? J Transplant. (2018) 2018:8316860. 10.1155/2018/8316860
    1. Viglietti D, Loupy A, Aubert O, Bestard O, Duong Van Huyen JP, Taupin JL, et al. . Dynamic prognostic score to predict kidney allograft survival in patients with antibody-mediated rejection. J Am Soc Nephrol. (2018) 29:606–619. 10.1681/ASN.2017070749
    1. Aubert O, Loupy A, Hidalgo L, Duong van Huyen JP, Higgins S, Viglietti D, et al. . Antibody-mediated rejection due to preexisting versus de novo donor-specific antibodies in kidney allograft recipients. J Am Soc Nephrol. (2017) 28:1912–23. 10.1681/ASN.2016070797
    1. Courant M, Visentin J, Linares G, Dubois V, Lepreux S, Guidicelli G, et al. . The disappointing contribution of anti-human leukocyte antigen donor-specific antibodies characteristics for predicting allograft loss. Nephrol Dial Transplant. (2018) 33:1853–63. 10.1093/ndt/gfy088
    1. Yell M, Muth BL, Kaufman DB, Djamali A, Ellis TM. C1q binding activity of de novo donor-specific hla antibodies in renal transplant recipients with and without antibody-mediated rejection. Transplantation. (2015) 99:1151–5. 10.1097/TP.0000000000000699

Source: PubMed

3
Suscribir