A novel approach reveals that HLA class 1 single antigen bead-signatures provide a means of high-accuracy pre-transplant risk assessment of acute cellular rejection in renal transplantation

Nicole Wittenbrink, Sabrina Herrmann, Arturo Blazquez-Navarro, Chris Bauer, Eric Lindberg, Kerstin Wolk, Robert Sabat, Petra Reinke, Birgit Sawitzki, Oliver Thomusch, Christian Hugo, Nina Babel, Harald Seitz, Michal Or-Guil, Nicole Wittenbrink, Sabrina Herrmann, Arturo Blazquez-Navarro, Chris Bauer, Eric Lindberg, Kerstin Wolk, Robert Sabat, Petra Reinke, Birgit Sawitzki, Oliver Thomusch, Christian Hugo, Nina Babel, Harald Seitz, Michal Or-Guil

Abstract

Background: Acute cellular rejection (ACR) is associated with complications after kidney transplantation, such as graft dysfunction and graft loss. Early risk assessment is therefore critical for the improvement of transplantation outcomes. In this work, we retrospectively analyzed a pre-transplant HLA antigen bead assay data set that was acquired by the e:KID consortium as part of a systems medicine approach.

Results: The data set included single antigen bead (SAB) reactivity profiles of 52 low-risk graft recipients (negative complement dependent cytotoxicity crossmatch, PRA < 30%) who showed detectable pre-transplant anti-HLA 1 antibodies. To assess whether the reactivity profiles provide a means for ACR risk assessment, we established a novel approach which differs from standard approaches in two aspects: the use of quantitative continuous data and the use of a multiparameter classification method. Remarkably, it achieved significant prediction of the 38 graft recipients who experienced ACR with a balanced accuracy of 82.7% (sensitivity = 76.5%, specificity = 88.9%).

Conclusions: The resultant classifier achieved one of the highest prediction accuracies in the literature for pre-transplant risk assessment of ACR. Importantly, it can facilitate risk assessment in non-sensitized patients who lack donor-specific antibodies. As the classifier is based on continuous data and includes weak signals, our results emphasize that not only strong but also weak binding interactions of antibodies and HLA 1 antigens contain predictive information.

Trial registration: ClinicalTrials.gov NCT00724022 . Retrospectively registered July 2008.

Keywords: Acute cellular rejection; Anti-HLA-1 antibodies; Immune signatures; Machine learning; Pre-transplantation risk assessment; Renal transplantation; Single HLA antigen bead assay.

Conflict of interest statement

Authors’ information

The authors are part of the e:KID consortium, an interdisciplinary group which has as its main aim the development and establishment a systems medicine approach to personalized immunosuppressive treatment at an early stage after kidney transplantation. The consortium’s backgrounds areas include expertise in bioinformatics and machine learning (NW, ABN, CB, EL and MO), HLA antibody experimental analysis and immunology (NW, SH, KW, RS, BS, HS and MO), and the clinic of renal transplantation (PR, OT, CH and NB).

Ethics approval and consent to participate

The study was carried out in compliance with the Declaration of Helsinki and Good Clinical Practice. All participants provided written informed consent prior to inclusion into the study. The trial was approved by the Ethics Committee of the Universitätsklinikum Carl Gustav Carus Dresden. The trial is registered with ClinicalTrials.gov, number NCT00724022 (https://ichgcp.net/clinical-trials-registry/NCT00724022). Date of registration was July 2008 (retrospectively registered), date of enrollment was June 2008.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Predictive performance of the multiparameter ACR risk assessment classifier based on rank-normalized continuous pre-transplant HLA-1 antibody reactivity profiles. a Output of the classifiers decision function for each patient. The decision threshold is indicated by a dashed horizontal line. Patients with a decision value > 0 are classified as ACR, patients with a decision value < 0 are classified as control. Colors indicate whether patients tested positive (black) or negative (grey) for the presence of serum HLA-1 antibodies during MAB screening. b ROC curve of the multiparameter classifier
Fig. 2
Fig. 2
Conventional MFI-thresholded binary MAB screening data do not allow for pre-transplant risk assessment of ACR. Illustrated are the results of the MAB screening data of the cohort (117 graft recipients, 63 ACR + 54 controls; for demographics and clinical characteristics, see Additional file 5: Table S2).). Analyses were carried out on MFI-thresholded binary HLA MAB screening data (conventional approach); according to Fisher’s exact test, differences with respect to the prevalence of HLA antibodies are not significant (p > 0.05)

References

    1. Djamali A, Kaufman DB, Ellis TM, Zhong W, Matas A, Samaniego M. Diagnosis and management of antibody-mediated rejection: current status and novel approaches. Am J Transplant. 2014;14(2):255–271.
    1. Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B. 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(3):378–383.
    1. Meier-Kriesche HU, Schold JD, Kaplan B. Long-term renal allograft survival: have we made significant progress or is it time to rethink our analytic and therapeutic strategies? Am J Transplant. 2004;4(8):1289–1295.
    1. Thomusch O, Wiesener M, Opgenoorth M, Pascher A, Woitas RP, Witzke O, et al. Rabbit-ATG or basiliximab induction for rapid steroid withdrawal after renal transplantation (harmony): an open-label, multicentre, randomised controlled trial. Lancet. 2016;388:3006–3016.
    1. Ciancio G, Burke GW, Gaynor JJ, Carreno MR, Cirocco RE, Mathew JM, et al. A randomized trial of three renal transplant induction antibodies: early comparison of tacrolimus, mycophenolate Mofetil, and steroid dosing, and newer immune-Monitoring1. Transplantation. 2005;80(4):457–465.
    1. Ekberg H, Tedesco-Silva H, Demirbas A, Vítko Š, Nashan B, Gürkan A, et al. Reduced exposure to Calcineurin inhibitors in renal transplantation. N Engl J Med. 2007;357(25):2562–2575.
    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(3):527–535.
    1. Roberts DM, Jiang SH, Chadban SJ. The treatment of acute antibody-mediated rejection in kidney transplant recipients—a systematic review. Transp J. 2012;94(8):775–783.
    1. Magil AB. Monocytes/macrophages in renal allograft rejection. Transplant Rev. 2009;23(4):199–208.
    1. Becker LE, Morath C, Suesal C. Immune mechanisms of acute and chronic rejection. Clin Biochem. 2016;49(4–5):320–323.
    1. Suthanthiran M, Schwartz JE, Ding R, Abecassis M, Dadhania D, Samstein B, Knechtle SJ, et al. Urinary-cell mRNA profile and acute cellular rejection in kidney allografts. N Engl J Med. 2013;369:20–31.
    1. Afaneh C, Muthukumar T, Lubetzky M, Ding R, Snopkowski C, Sharma VK, et al. Urinary cell levels of mRNA for OX40, OX40L, PD-1, PD-L1, or PD-L2 and acute rejection of human renal allografts. Transplantation. 2010;90(12):1381–1387.
    1. Hricik DE, Nickerson P, Formica RN, Poggio ED, Rush D, Newell KA, et al. Multicenter validation of urinary CXCL9 as a risk-stratifying biomarker for kidney transplant injury. Am J Transplant. 2013;13(10):2634–2644.
    1. Roedder S, Sigdel T, Salomonis N, Hsieh S, Dai H, Bestard O, et al. The kSORT assay to detect renal transplant patients at high risk for acute rejection: results of the multicenter AART study. PLoS Med. 2014;11(11):e1001759.
    1. Wang JN, Zhou Y, Zhu TY, Wang X, Guo YL. Prediction of acute cellular renal allograft rejection by urinary metabolomics using MALDI-FTMS. J Proteome Res. 2008;7(8):3597–3601.
    1. Reeve J, Einecke G, Mengel M, Sis B, Kayser N, Kaplan B, et al. Diagnosing rejection in renal transplants: a comparison of molecular- and histopathology-based approaches. Am J Transplant. 2009;9(8):1802–1810.
    1. Desvaux D, Schwarzinger M, Pastural M, Baron C, Abtahi M, Berrehar F, et al. Molecular diagnosis of renal-allograft rejection: correlation with histopathologic evaluation and antirejection-therapy resistance. Transplantation. 2004;78(5):647–653.
    1. Ting YT, Coates PT, Marti H-P, Dunn AC, Parker RM, Pickering JW, et al. Urinary soluble HLA-DR is a potential biomarker for acute renal transplant rejection. Transplantation. 2010;89(9):1071–1078.
    1. Poggio ED, Augustine JJ, Clemente M, Danzig JM, Volokh N, Zand MS, et al. Pretransplant cellular Alloimmunity as assessed by a panel of reactive T cells assay correlates with acute renal graft rejection. Transplantation. 2007;83(7):847–852.
    1. Simon T, Opelz G, Wiesel M, Pelzl S, Ott RC, Süsal C. Serial peripheral blood Interleukin-18 and perforin gene expression measurements for prediction of acute kidney graft rejection. Am J Transplant. 2003;3(1121–1127):1589–1595.
    1. Hauser IA. Prediction of acute renal allograft rejection by urinary Monokine induced by IFN- (MIG) J Am Soc Nephrol. 2005;16(6):1849–1858.
    1. Mancebo E, Castro MJ, Allende LM, Talayero P, Brunet M, Millán O, et al. High proportion of CD95+ and CD38+ in cultured CD8+ T cells predicts acute rejection and infection, respectively, in kidney recipients. Transpl Immunol. 2016;34:33–41.
    1. Dong W, Shunliang Y, Weizhen W, Qinghua W, Zhangxin Z, Jianming T, et al. Prediction of acute renal allograft rejection in early post-transplantation period by soluble CD30. Transpl Immunol. 2006;16(1):41–45.
    1. Malheiro J, Tafulo S, Dias L, Martins LS, Fonseca I, Beirão I, et al. Analysis of preformed donor-specific anti-HLA antibodies characteristics for prediction of antibody-mediated rejection in kidney transplantation. Transpl Immunol. 2015;32(2):66–71.
    1. Vlad G, Ho EK, Vasilescu ER, Colovai AI, Stokes MB, Markowitz GS, et al. Relevance of different antibody detection methods for the prediction of antibody-mediated rejection and deceased-donor kidney allograft survival. Hum Immunol. 2009;70(8):589–594.
    1. Riethmüller S, Ferrari-Lacraz S, Müller MK, Raptis DA, Hadaya K, Rüsi B, et al. Donor-specific antibody levels and three generations of crossmatches to predict antibody-mediated rejection in kidney transplantation. Transp J. 2010;90(2):160–167.
    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(8):1398–1406.
    1. Song EY, Lee YJ, Hyun J, Kim YS, Ahn C, Ha J, et al. Clinical relevance of pretransplant HLA class II donor-specific antibodies in renal transplantation patients with negative T-cell cytotoxicity crossmatches. Ann Lab Med. 2012;32(2):139–144.
    1. Salvadé I, Aubert V, Venetz JP, Golshayan D, Saouli AC, Matter M, et al. Clinically-relevant threshold of preformed donor-specific anti-HLA antibodies in kidney transplantation. Hum Immunol. 2016;77(6):483–489.
    1. Shaikhina T, Lowe D, Daga S, Briggs D, Higgins R, Khovanova N. Decision tree and random forest models for outcome prediction in antibody incompatible kidney transplantation. Biomedical Signal Processing Control. 2017; In press. DOI: 10.1016/j.bspc.2017.01.012.
    1. Ho EK, Vasilescu ER, Colovai AI, Stokes MB, Hallar M, Markowitz GS, et al. Sensitivity, specificity and clinical relevance of different cross-matching assays in deceased-donor renal transplantation. Transpl Immunol. 2008;20(1–2):61–67.
    1. Kannabhiran D, Lee J, Schwartz JE, Friedlander R, Aull M, Muthukumar T, et al. Characteristics of circulating donor human leukocyte antigen-specific immunoglobulin G antibodies predictive of acute antibody-mediated rejection and kidney allograft failure. Transplantation. 2015;99(6):1156–1164.
    1. Poggio ED, Clemente M, Hricik DE, Heeger PS. Panel of reactive T cells as a measurement of primed cellular Alloimmunity in kidney transplant candidates. J Am Soc Nephrol. 2006;17:564–572.
    1. Vondran FWR, Timrott K, Kollrich S, Steinhoff AK, Kaltenborn A, Schrem H, et al. Pre-transplant immune state defined by serum markers and alloreactivity predicts acute rejection after living donor kidney transplantation. Clin Transpl. 2014;28(9):968–979.
    1. Nafar M, Farrokhi F, Vaezi M, Entezari AE, Pour-Reza-Gholi F, Firoozan A, et al. Pre-transplant and post-transplant soluble CD30 for prediction and diagnosis of acute kidney allograft rejection. Int Urol Nephrol. 2009;41(3):687–693.
    1. Hughes AK, Cichacz Z, Scheck A, Coons SW, Johnston SA, Stafford P. Immunosignaturing can detect products from molecular markers in brain cancer. PLoS One. 2012;7(7):e40201.
    1. Restrepo L, Stafford P, Johnston SA. Feasibility of an early Alzheimer’s disease immunosignature diagnostic test. J Neuroimmunol. 2013;254(1–2):154–160.
    1. Stafford P, Cichacz Z, Woodbury NW, Johnston SA. Immunosignature system for diagnosis of cancer. Proc Natl Acad Sci U S A. 2014;111(30):E3072–E3080.
    1. Legutki JB, Johnston SA. Immunosignatures can predict vaccine efficacy. Proc Natl Acad Sci U S A. 2013;110(46):18614–18619.
    1. Cascalho MI, Chen BJ, Kain M, Platt JL. The paradoxical functions of B cells and antibodies in transplantation. J Immunol. 2013;190(3):875–879.
    1. Randhawa P. T-cell-mediated rejection of the kidney in the era of donor-specific antibodies: diagnostic challenges and clinical significance. Curr Opin Organ Transplant. 2015;20(3):325–332.
    1. Blazquez-Navarro A, Schachtner T, Stervbo U, Sefrin A, Stein M, Westhoff TH, et al. Differential T cell response against BK virus regulatory and structural antigens: a viral dynamics modelling approach. PLoS Comput Biol. 2018;14(5):1–20.
    1. Blazquez-Navarro A, Dang-Heine C, Wittenbrink N, Bauer C, Wolk K, Sabat R, et al. BKV, CMV, and EBV interactions and their effect on graft function one year post-renal transplantation: results from a large multi-Centre study. EBioMedicine. 2018;34:113–121.
    1. Sullivan HC, Liwski RS, Bray RA, Gebel HM. The road to HLA antibody evaluation: do not rely on MFI. Am J Transplant. 2017;XX:1–7.
    1. Konvalinka A, Tinckam K. Utility of HLA antibody testing in kidney transplantation. J Am Soc Nephrol. 2015;26(7):1489–1502.
    1. Fairchild PJ, Wraith DC. Lowering the tone: mechanisms of immunodominance among epitopes with low affinity for MHC. Immunol Today. 1996;17(2):80–85.
    1. Haywood AM. Virus receptors: binding, adhesion strengthening, and changes in viral structure. J Virol. 1994;68(1):1–5.
    1. Hakomori S. Structure and function of Sphingoglycolipids in transmembrane Signalling and cell-cell interactions. Biochem Soc Trans. 1993;21(3):583–595.
    1. van der Merwe PA, Brown MH, Davis SJ, Barclay AN. Affinity and kinetic analysis of the interaction of the cell adhesion molecules rat CD2 and CD48. EMBO J. 1993;12(13):4945–4954.
    1. Reilly PL, Woska JR, Jeanfavre DD, McNally E, Rothlein R, Bormann BJ. The native structure of intercellular adhesion molecule-1 (ICAM-1) is a dimer. Correlation with binding to LFA-1. J Immunol. 1995;155(2):529–532.
    1. Hage DS. Affinity chromatography methods and protocols. 2000. Weak affinity chromatography; pp. 7–23.
    1. Szatmary P, Jones J, Hammad A, Middleton D. Impact of sensitivity of human leucocyte antigen antibody detection by Luminex technology on graft loss at 1 year. Clin Kidney J. 2013;6(3):283–286.
    1. Thomas KA, Valenzuela NM, Reed EF. The perfect storm: HLA antibodies, complement, FcγRs and endothelium in transplant rejection. Trends Mol Med. 2015;21(5):319–329.
    1. Pike R, Thomas N, Workman S, Ambrose L, Guzman D, Sivakumaran S, et al. PD1-expressing T cell subsets modify the rejection risk in renal transplant patients. Front Immunol. 2016;7:126.
    1. Zhang Q, Liu YF, Su ZX, Shi LP, Chen YH. Serum fractalkine and interferon-gamma inducible protein-10 concentrations are early detection markers for acute renal allograft rejection. Transplant Proc. 2014;46(5):1420–1425.
    1. Solez K, Colvin RB, Racusen LC, Sis B, Halloran PF, Birk PE, et al. Banff ‘05 meeting report: differential diagnosis of chronic allograft injury and elimination of chronic allograft nephropathy ('CAN’) Am J Transplant. 2007;7(3):518–526.
    1. Hochreiter S, Obermayer K. Support vector machines for dyadic data. Neural Comput. 2006;18(6):1472–1510.
    1. Molinaro AM, Simon R, Pfeiffer RM. Prediction error estimation: a comparison of resampling methods. Bioinformatics. 2005;21(15):3301–3307.
    1. Mason SJ, Graham NE. Areas beneath the relative operating characteristics (ROC) and relative operating levels (ROL) curves: statistical significance and interpretation. Q J R Meteorol Soc. 2002;128(584):2145–2166.

Source: PubMed

3
Se inscrever