Anti-Phospholipase A2 Receptor Antibody Titer Predicts Post-Rituximab Outcome of Membranous Nephropathy

Piero Ruggenenti, Hanna Debiec, Barbara Ruggiero, Antonietta Chianca, Timothee Pellé, Flavio Gaspari, Flavio Suardi, Elena Gagliardini, Silvia Orisio, Ariela Benigni, Pierre Ronco, Giuseppe Remuzzi, Piero Ruggenenti, Hanna Debiec, Barbara Ruggiero, Antonietta Chianca, Timothee Pellé, Flavio Gaspari, Flavio Suardi, Elena Gagliardini, Silvia Orisio, Ariela Benigni, Pierre Ronco, Giuseppe Remuzzi

Abstract

Rituximab induces nephrotic syndrome (NS) remission in two-thirds of patients with primary membranous nephropathy (MN), even after other treatments have failed. To assess the relationships among treatment effect, circulating nephritogenic anti-phospholipase A2 receptor (anti-PLA2R) autoantibodies and genetic polymorphisms predisposing to antibody production we serially monitored 24-hour proteinuria and antibody titer in patients with primary MN and long-lasting NS consenting to rituximab (375 mg/m(2)) therapy and genetic analyses. Over a median (range) follow-up of 30.8 (6.0-145.4) months, 84 of 132 rituximab-treated patients achieved complete or partial NS remission (primary end point), and 25 relapsed after remission. Outcomes of patients with or without detectable anti-PLA2R antibodies at baseline were similar. Among the 81 patients with antibodies, lower anti-PLA2R antibody titer at baseline (P=0.001) and full antibody depletion 6 months post-rituximab (hazard ratio [HR], 7.90; 95% confidence interval [95% CI], 2.54 to 24.60; P<0.001) strongly predicted remission. All 25 complete remissions were preceded by complete anti-PLA2R antibody depletion. On average, 50% anti-PLA2R titer reduction preceded equivalent proteinuria reduction by 10 months. Re-emergence of circulating antibodies predicted disease relapse (HR, 6.54; 95% CI, 1.57 to 27.40; P=0.01), whereas initial complete remission protected from the event (HR, 6.63; 95% CI, 2.37 to 18.53; P<0.001). Eighteen patients achieved persistent antibody depletion and complete remission and never relapsed. Outcome was independent of PLA2R1 and HLA-DQA1 polymorphisms and of previous immunosuppressive treatment. Therefore, assessing circulating anti-PLA2R autoantibodies and proteinuria may help in monitoring disease activity and guiding personalized rituximab therapy in nephrotic patients with primary MN.

Keywords: glomerulonephritis; membranous nephropathy; nephrotic syndrome; polymorphisms; primary; proteinuria.

Copyright © 2015 by the American Society of Nephrology.

Figures

Figure 1.
Figure 1.
Outcome of the 132 study patients who were available for analyses after rituximab administration. The flow chart shows how many of the 132 patients were or were not screened for anti-PLA2R antibodies and in how many of screened patients, the antibodies were or were not detectable. For each subgroup the flow chart provides detail on patients achieving or not achieving remission after rituximab therapy, progressing or not progressing to a relapse after initial remission, who are completing active follow-up after rituximab administration or are planned for rituximab therapy.
Figure 2.
Figure 2.
Kaplan–Meier curves for the proportion of participants with primary MN who progressed to the combined end point of complete or partial remission (left panel) or complete remission considered as a single end point (right panel) according to treatment with the four-dose or the B cell–driven regimen. In the unadjusted analysis of complete remission, we included a time-varying covariate to account for the unmet proportional hazard assumption. Progression to both end points did not differ appreciably between groups. Adjustments were by sex and log-transformed serum creatinine, and proteinuria. 95% CI, 95% confidence interval; HR, hazard ratio.
Figure 3.
Figure 3.
Kaplan–Meier curves for the proportion of participants with primary MN who progressed to the combined end point of complete or partial remission (left panel) or complete remission considered as a single end point (right panel) in three subgroups with or without detectable anti-PLA2R autoantibodies or without anti-PLA2R antibody evaluations at baseline. Progression to both end points did not differ appreciably among the groups. The upper and lower HRs refer to not available and the detectable group versus the undetectable group (reference), respectively. 95% CI, 95% confidence interval; HR, hazard ratio.
Figure 4.
Figure 4.
Kaplan–Meier curves for the proportion of participants with primary MN and detectable antibodies at baseline who achieved the combined end point of complete or partial remission (left panel) or complete remission considered as a single end point (right panel) in three tertiles of baseline anti-PLA2R autoantibody titer. The probability of achieving both end points progressively decreased from the lowest to the middle and the highest tertile (reference). The upper and lower HRs refer to lowest and middle tertiles versus highest tertile (reference), respectively. 95% CI, 95% confidence interval; HR, hazard ratio.
Figure 5.
Figure 5.
Mean±SEM percent changes in proteinuria, serum albumin, and anti-PLA2R autoantibody levels and circulating CD20 cell counts at different time points after rituximab administration compared with baseline (month 0). °P<0.05, *P≤0.01, ***P≤0.001 (all versus baseline).
Figure 6.
Figure 6.
Mean±SEM percent changes in proteinuria (highest panel), circulating CD20 cell counts (middle panel), and serum anti-PLA2R autoantibody levels (lowest panel) at different time points after rituximab administration compared with baseline (month 0) in patients who progressed to the combined end point of complete or partial remission, progressed to complete remission considered as a single end point, or achieved no remission. °P<0.05, *P≤0.01, **P≤0.001, ***P≤0.0001 (all versus no remission).
Figure 7.
Figure 7.
Kaplan–Meier curves for the proportion of participants with primary MN and detectable antibodies at baseline who achieved the combined end point of complete or partial remission (left panel) or complete remission considered as a single end point (right panel) according to depletion of anti-PLA2R autoantibodies over 6 months after rituximab administration. Autoantibody depletion significantly predicts an increased probability of achieving both end points. HRs were adjusted for sex and log-transformed serum creatinine, proteinuria, and anti-PLA2R autoantibody titer. CI, confidence interval; HR, hazard ratio.
Figure 8.
Figure 8.
Kaplan–Meier curves for the proportion of participants with primary MN with a relapse of the NS after initial complete or partial remission considered according to the four-dose or B cell–driven rituximab protocol. Risk of relapse is not associated with the treatment regimen even after adjusting for sex and log-transformed serum creatinine and proteinuria. HRs refer to B cell–driven group versus four-dose group (reference). 95% CI, 95% confidence interval; HR, hazard ratio.
Figure 9.
Figure 9.
Kaplan–Meier curves for the proportion of participants with primary MN with a relapse of the NS after initial remission considered according to previous complete or partial remission. Previous complete remission is associated with a significant protective effect against subsequent disease relapse even after adjusting for age, sex, and log-transformed proteinuria. 95% CI, 95% confidence interval; HR, hazard ratio.
Figure 10.
Figure 10.
Kaplan–Meier curves for the proportion of participants with primary MN with a relapse of the NS after initial complete or partial remission considered according to previous anti-PLA2R autoantibody titer increase/re-emergence after initial rituximab-induced reduction/depletion (left panel) or re-emergence only into circulation after initial depletion (right panel). In both cases, titer increase or antibody re-emergence are associated with a significant excess risk of subsequent disease relapse, even after adjusting for other risk factors (age). 95% CI, 95% confidence interval; HR, hazard ratio.

References

    1. Remuzzi G, Chiurchiu C, Abbate M, Brusegan V, Bontempelli M, Ruggenenti P: Rituximab for idiopathic membranous nephropathy. Lancet 360: 923–924, 2002
    1. Bomback AS, Derebail VK, McGregor JG, Kshirsagar AV, Falk RJ, Nachman PH: Rituximab therapy for membranous nephropathy: A systematic review. Clin J Am Soc Nephrol 4: 734–744, 2009
    1. Cravedi P, Sghirlanzoni MC, Marasà M, Salerno A, Remuzzi G, Ruggenenti P: Efficacy and safety of rituximab second-line therapy for membranous nephropathy: A prospective, matched-cohort study. Am J Nephrol 33: 461–468, 2011
    1. Segarra A, Praga M, Ramos N, Polanco N, Cargol I, Gutierrez-Solis E, Gomez MR, Montoro B, Camps J: Successful treatment of membranous glomerulonephritis with rituximab in calcineurin inhibitor-dependent patients. Clin J Am Soc Nephrol 4: 1083–1088, 2009
    1. Ruggenenti P, Cravedi P, Chianca A, Perna A, Ruggiero B, Gaspari F, Rambaldi A, Marasà M, Remuzzi G: Rituximab in idiopathic membranous nephropathy. J Am Soc Nephrol 23: 1416–1425, 2012
    1. Glassock RJ: Human idiopathic membranous nephropathy--a mystery solved? N Engl J Med 361: 81–83, 2009
    1. Ruggenenti P, Cravedi P, Remuzzi G: Latest treatment strategies for membranous nephropathy. Expert Opin Pharmacother 8: 3159–3171, 2007
    1. Debiec H, Guigonis V, Mougenot B, Decobert F, Haymann JP, Bensman A, Deschênes G, Ronco PM: Antenatal membranous glomerulonephritis due to anti-neutral endopeptidase antibodies. N Engl J Med 346: 2053–2060, 2002
    1. Beck LH, Jr, Bonegio RG, Lambeau G, Beck DM, Powell DW, Cummins TD, Klein JB, Salant DJ: M-type phospholipase A2 receptor as target antigen in idiopathic membranous nephropathy. N Engl J Med 361: 11–21, 2009
    1. Cravedi P, Ruggenenti P, Remuzzi G: Circulating anti-PLA2R autoantibodies to monitor immunological activity in membranous nephropathy. J Am Soc Nephrol 22: 1400–1402, 2011
    1. Stanescu HC, Arcos-Burgos M, Medlar A, Bockenhauer D, Kottgen A, Dragomirescu L, Voinescu C, Patel N, Pearce K, Hubank M, Stephens HA, Laundy V, Padmanabhan S, Zawadzka A, Hofstra JM, Coenen MJ, den Heijer M, Kiemeney LA, Bacq-Daian D, Stengel B, Powis SH, Brenchley P, Feehally J, Rees AJ, Debiec H, Wetzels JF, Ronco P, Mathieson PW, Kleta R: Risk HLA-DQA1 and PLA(2)R1 alleles in idiopathic membranous nephropathy. N Engl J Med 364: 616–626, 2011
    1. Bullich G, Ballarín J, Oliver A, Ayasreh N, Silva I, Santín S, Díaz-Encarnación MM, Torra R, Ars E: HLA-DQA1 and PLA2R1 polymorphisms and risk of idiopathic membranous nephropathy. Clin J Am Soc Nephrol 9: 335–343, 2014
    1. Lv J, Hou W, Zhou X, Liu G, Zhou F, Zhao N, Hou P, Zhao M, Zhang H: Interaction between PLA2R1 and HLA-DQA1 variants associates with anti-PLA2R antibodies and membranous nephropathy. J Am Soc Nephrol 24: 1323–1329, 2013
    1. Debiec H, Ronco P: Immunopathogenesis of membranous nephropathy: An update. Semin Immunopathol 36: 381–397, 2014
    1. Cravedi P, Ruggenenti P, Sghirlanzoni MC, Remuzzi G: Titrating rituximab to circulating B cells to optimize lymphocytolytic therapy in idiopathic membranous nephropathy. Clin J Am Soc Nephrol 2: 932–937, 2007
    1. Kanigicherla D, Gummadova J, McKenzie EA, Roberts SA, Harris S, Nikam M, Poulton K, McWilliam L, Short CD, Venning M, Brenchley PE: Anti-PLA2R antibodies measured by ELISA predict long-term outcome in a prevalent population of patients with idiopathic membranous nephropathy. Kidney Int 83: 940–948, 2013
    1. Hoxha E, Thiele I, Zahner G, Panzer U, Harendza S, Stahl RA: Phospholipase A2 receptor autoantibodies and clinical outcome in patients with primary membranous nephropathy. J Am Soc Nephrol 25: 1357–1366, 2014
    1. Hofstra JM, Debiec H, Short CD, Pellé T, Kleta R, Mathieson PW, Ronco P, Brenchley PE, Wetzels JF: Antiphospholipase A2 receptor antibody titer and subclass in idiopathic membranous nephropathy. J Am Soc Nephrol 23: 1735–1743, 2012
    1. Hofstra JM, Beck LH, Jr, Beck DM, Wetzels JF, Salant DJ: Anti-phospholipase A₂ receptor antibodies correlate with clinical status in idiopathic membranous nephropathy. Clin J Am Soc Nephrol 6: 1286–1291, 2011
    1. Beck LH, Jr, Fervenza FC, Beck DM, Bonegio RG, Malik FA, Erickson SB, Cosio FG, Cattran DC, Salant DJ: Rituximab-induced depletion of anti-PLA2R autoantibodies predicts response in membranous nephropathy. J Am Soc Nephrol 22: 1543–1550, 2011
    1. Penny MJ, Boyd RA, Hall BM: Mycophenolate mofetil prevents the induction of active Heymann nephritis: Association with Th2 cytokine inhibition. J Am Soc Nephrol 9: 2272–2282, 1998
    1. Coiffier B, Pfreundschuh M, Stahel R, Vose J, Zinzani PL: Aggressive lymphoma: Improving treatment outcome with rituximab. Anticancer Drugs 13[Suppl 2]: S43–S50, 2002
    1. Ruggenenti P, Ruggiero B, Cravedi P, Vivarelli M, Massella L, Marasà M, Chianca A, Rubis N, Ene-Iordache B, Rudnicki M, Pollastro RM, Capasso G, Pisani A, Pennesi M, Emma F, Remuzzi G, Rituximab in Nephrotic Syndrome of Steroid-Dependent or Frequently Relapsing Minimal Change Disease Or Focal Segmental Glomerulosclerosis (NEMO) Study Group : Rituximab in steroid-dependent or frequently relapsing idiopathic nephrotic syndrome. J Am Soc Nephrol 25: 850–863, 2014
    1. Cambridge G, Isenberg DA, Edwards JC, Leandro MJ, Migone TS, Teodorescu M, Stohl W: B cell depletion therapy in systemic lupus erythematosus: Relationships among serum B lymphocyte stimulator levels, autoantibody profile and clinical response. Ann Rheum Dis 67: 1011–1016, 2008
    1. Teng YK, Wheater G, Hogan VE, Stocks P, Levarht EW, Huizinga TW, Toes RE, van Laar JM: Induction of long-term B-cell depletion in refractory rheumatoid arthritis patients preferentially affects autoreactive more than protective humoral immunity. Arthritis Res Ther 14: R57, 2012
    1. Fervenza FC, Abraham RS, Erickson SB, Irazabal MV, Eirin A, Specks U, Nachman PH, Bergstralh EJ, Leung N, Cosio FG, Hogan MC, Dillon JJ, Hickson LJ, Li X, Cattran DC, Mayo Nephrology Collaborative Group : Rituximab therapy in idiopathic membranous nephropathy: A 2-year study. Clin J Am Soc Nephrol 5: 2188–2198, 2010
    1. Fresquet M, Jowitt TA, Gummadova J, Collins R, O’Cualain R, McKenzie EA, Lennon R, Brenchley PE: Identification of a major epitope recognized by PLA2R autoantibodies in primary membranous nephropathy [published online ahead of print October 6, 2014]. J Am Soc Nephrol
    1. Kim S, Chin HJ, Na KY, Kim S, Oh J, Chung W, Noh JW, Lee YK, Cho JT, Lee EK, Chae DW, Progressive Renal Disease and Medical Informatics and Genomics Research (PREMIER) members : Single nucleotide polymorphisms in the phospholipase A2 receptor gene are associated with genetic susceptibility to idiopathic membranous nephropathy. Nephron Clin Pract 117: c253–c258, 2011
    1. Bresin E, Gastoldi S, Daina E, Belotti D, Pogliani E, Perseghin P, Scalzulli PR, Paolini R, Marcenò R, Remuzzi G, Galbusera M: Rituximab as pre-emptive treatment in patients with thrombotic thrombocytopenic purpura and evidence of anti-ADAMTS13 autoantibodies. Thromb Haemost 101: 233–238, 2009
    1. Royston P, Sauerbrei W: Multivariable model-building. A pragmatic approach to regression analysis based on fractional polynomials for modelling continuous variables, John Wiley & Sons, 2008
    1. Royston P, Sauerbrei W, Becher H: Modelling continuous exposures with a ‘spike’ at zero: A new procedure based on fractional polynomials. Stat Med 29: 1219–1227, 2010
    1. Becher H, Lorenz E, Royston P, Sauerbrei W: Analysing covariates with spike at zero: A modified FP procedure and conceptual issues. Biom J 54: 686–700, 2012

Source: PubMed

3
Abonner