Management of Lupus Nephritis

Farah Tamirou, Frédéric A Houssiau, Farah Tamirou, Frédéric A Houssiau

Abstract

Lupus nephritis (LN) is a frequent and severe manifestation of systemic lupus erythematosus. The main goal of the management of LN is to avoid chronic kidney disease (CKD). Current treatment strategies remain unsatisfactory in terms of complete renal response, prevention of relapses, CKD, and progression to end-stage kidney disease. To improve the prognosis of LN, recent data suggest that we should (i) modify our treat-to-target approach by including, in addition to a clinical target, a pathological target and (ii) switch from conventional sequential therapy to combination therapy. Here, we also review the results of recent controlled randomized trials.

Keywords: combination therapy; lupus nephritis; repeat kidney biopsy; treat-to-target approach.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
REBIOLUP flowchart.

References

    1. Rovin B.H., Stillman I. Kidney. In: Lahita R., editor. Systemic Lupus Erythematosus. 5th ed. Academic Press; San Diego, CA, USA: 2011. pp. 769–814.
    1. Wang H., Ren Y.L., Chang J., Gu L., Sun L.Y. A systematic review and meta-analysis of prevalence of biopsy-proven lupus nephritis. Arch. Rheumatol. 2017;33:17–25. doi: 10.5606/ArchRheumatol.2017.6127.
    1. Seligman V.A., Suarez C., Lum R., Inda S.E., Lin D., Li H., Olson J.L., Seldin M.F., Criswell L.A. The Fcgamma receptor IIIA-158F allele is a major risk factor for the development of lupus nephritis among Caucasians but not non-Caucasians. Arthritis Rheum. 2001;44:618–625. doi: 10.1002/1529-0131(200103)44:3<618::AID-ANR110>;2-R.
    1. Lech M., Anders H.J. The pathogenesis of lupus nephritis. J. Am. Soc. Nephrol. 2013;24:1357–1366. doi: 10.1681/ASN.2013010026.
    1. Tojo T., Friou G.J. Lupus nephritis: Varying complement-fixing properties of immunoglobulin G antibodies to antigens of cell nuclei. Science. 1968;161:904–906. doi: 10.1126/science.161.3844.904.
    1. Kramers C., Hylkema M.N., van Bruggen M.C., van de Lagemaat R., Dijkman H.B., Assmann K.J., Smeenk R.J., Berden J.H. Anti-nucleosome antibodies complexed to nucleosomal antigens show anti-DNA reactivity and bind to rat glomerular basement membrane in vivo. J. Clin. Investig. 1994;94:568–577. doi: 10.1172/JCI117371.
    1. Mjelle J.E., Rekvig O.P., Van Der Vlag J., Fenton K.A. Nephritogenic antibodies bind in glomeruli through interaction with exposed chromatin fragments and not with renal cross-reactive antigens. Autoimmunity. 2011;44:373–383. doi: 10.3109/08916934.2010.541170.
    1. Hahn B.H. Antibodies to DNA. N. Engl. J. Med. 1998;338:1359–1368. doi: 10.1056/NEJM199805073381906.
    1. Weening J.J., D’Agati V.D., Schwartz M.M., Seshan S.V., Alpers C.E., Appel G.B., Balow J.E., Bruijn J.A., Cook T., Ferrario F., et al. The classification of glomerulonephritis in systemic lupus erythematosus revisited. Kidney Int. 2004;65:521–530. doi: 10.1111/j.1523-1755.2004.00443.x.
    1. Chang A., Henderson S.G., Brandt D., Liu N., Guttikonda R., Hsieh C., Kaverina N., Utset T.O., Meehan S.M., Quigg R.J., et al. In situ B cell-mediated immune responses and tubulointerstitial inflammation in human lupus nephritis. J. Immunol. 2011;186:1849–1860. doi: 10.4049/jimmunol.1001983.
    1. Maria N.I., Davidson A. Protecting the kidney in systemic lupus erythematosus: From diagnosis to therapy. Nat. Rev. Rheumatol. 2020;16:255–267. doi: 10.1038/s41584-020-0401-9.
    1. Christopher-Stine L., Siedner M., Lin J., Haas M., Parekh H., Petri M., Fine D.M. Renal biopsy in lupus patients with low levels of proteinuria. J. Rheumatol. 2007;34:332–335.
    1. De Rosa M., Rocha A.S., De Rosa G., Dubinsky D., Almaani S.J., Rovin B.H. Low-grade proteinuria does not exclude significant kidney injury in lupus nephritis. Kidney Int. Rep. 2020;5:1066–1068. doi: 10.1016/j.ekir.2020.04.005.
    1. Fiehn C., Hajjar Y., Mueller K., Waldherr R., Ho A.D., Andrassy K. Improved clinical outcome of lupus nephritis during the past decade: Importance of early diagnosis and treatment. Ann. Rheum. Dis. 2003;62:435–439. doi: 10.1136/ard.62.5.435.
    1. Singh A.K., Ucci A., Madias N.E. Predominant tubulointerstitial lupus nephritis. Am. J. Kidney Dis. 1996;27:273–278. doi: 10.1016/S0272-6386(96)90553-3.
    1. Leatherwood C., Speyer C.B., Feldman C.H., D’Silva K., Gómez-Puerta J.A., Hoover P.J., Waikar S.S., McMahon G.M., Rennke H.G., Costenbader K.H. Clinical characteristics and renal prognosis associated with interstitial fibrosis and tubular atrophy (IFTA) and vascular injury in lupus nephritis biopsies. Semin. Arthritis Rheum. 2019;49:396–404. doi: 10.1016/j.semarthrit.2019.06.002.
    1. Vandepapelière J., Aydin S., Cosyns J.P., Depresseux G., Jadoul M., Houssiau F.A. Prognosis of proliferative lupus nephritis subsets in the Louvain Lupus Nephritis inception Cohort. Lupus. 2014;23:159–165. doi: 10.1177/0961203313514623.
    1. Hsieh C., Chang A., Brandt D., Guttikonda R., Utset T.O., Clark M.R. Predicting outcomes of lupus nephritis with tubulointerstitial inflammation and scarring. Arthritis Care Res. 2011;63:865–874. doi: 10.1002/acr.20441.
    1. Bajema I.M., Wilhelmus S., Alpers C.E., Bruijn J.A., Colvin R.B., Cook H.T., D’Agati V.D., Ferrario F., Haas M., Jennette J.C., et al. Revision of the International Society of Nephrology/Renal Pathology Society classification for lupus nephritis: Clarification of definitions, and modified National Institutes of Health activity and chronicity indices. Kidney Int. 2018;93:789–796. doi: 10.1016/j.kint.2017.11.023.
    1. Fanouriakis A., Kostopoulou M., Cheema K., Anders H.J., Aringer M., Bajema I., Boletis J., Frangou E., Houssiau F.A., Hollis J., et al. 2019 Update of the Joint European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of lupus nephritis. Ann. Rheum. Dis. 2020;79:713–723. doi: 10.1136/annrheumdis-2020-216924.
    1. Houssiau F.A., Vasconcelos C., D’Cruz D., Sebastiani G.D., Garrido Ed E., Danieli M.G., Abramovicz D., Blockmans D., Mathieu A., Direskeneli H., et al. Immunosuppressive therapy in lupus nephritis: The Euro-Lupus Nephritis Trial, a randomized trial of low-dose versus high-dose intravenous cyclophosphamide. Arthritis Rheum. 2002;46:2121–2131. doi: 10.1002/art.10461.
    1. Austin H.A., III, Klippel J.H., Balow J.E., le Riche N.G., Steinberg A.D., Plotz P.H., Decker J.L. Therapy of lupus nephritis. Controlled trial of prednisone and cytotoxic drugs. N. Engl. J. Med. 1986;314:614–619. doi: 10.1056/NEJM198603063141004.
    1. Zeher M., Doria A., Lan J., Aroca G., Jayne D., Boletis I., Hiepe F., Prestele H., Bernhardt P., Amoura Z. Efficacy and safety of enteric-coated mycophenolate sodium in combination with two glucocorticoid regimens for the treatment of active lupus nephritis. Lupus. 2011;20:1484–1493. doi: 10.1177/0961203311418269.
    1. Lightstone L., Doria A., Wilson H., Ward F.L., Larosa M., Bargman J.M. Can we manage lupus nephritis without chronic corticosteroids administration? Autoimmun. Rev. 2018;17:4–10. doi: 10.1016/j.autrev.2017.11.002.
    1. Malvar A., Pirruccio P., Alberton V., Lococo B., Recalde C., Fazini B., Nagaraja H., Indrakanti D., Rovin B.H. Histologic versus clinical remission in proliferative lupus nephritis. Nephrol. Dial. Transplant. 2017;32:1338–1344. doi: 10.1093/ndt/gfv296.
    1. Dooley M.A., Jayne D., Ginzler E.M., Isenberg D., Olsen N.J., Wofsy D., Eitner F., Appel G.B., Contreras G., Lisk L., et al. Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N. Engl. J. Med. 2011;365:1886–1895. doi: 10.1056/NEJMoa1014460.
    1. El Hachmi M., Jadoul M., Lefèbvre C., Depresseux G., Houssiau F.A. Relapses of lupus nephritis: Incidence, risk factors, serology and impact on outcome. Lupus. 2003;12:692–696. doi: 10.1191/0961203303lu444oa.
    1. Parikh S.V., Nagaraja H.N., Hebert L., Rovin B.H. Renal flare as a predictor of incident and progressive CKD in patients with lupus nephritis. Clin. J. Am. Soc. Nephrol. 2014;9:279–284. doi: 10.2215/CJN.05040513.
    1. Houssiau F.A., Vasconcelos C., D’Cruz D., Sebastiani G.D., de Ramon Garrido E., Danieli M.G., Abramovicz D., Blockmans D., Cauli A., Direskeneli H., et al. The 10-year follow-up data of the Euro-Lupus Nephritis Trial comparing low-dose and high-dose intravenous cyclophosphamide. Ann. Rheum. Dis. 2010;69:61–64. doi: 10.1136/ard.2008.102533.
    1. Appel G.B., Contreras G., Dooley M.A., Ginzler E.M., Isenberg D., Jayne D., Li L.S., Mysler E., Sánchez-Guerrero J., Solomons N., et al. Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J. Am. Soc. Nephrol. 2009;20:1103–1112. doi: 10.1681/ASN.2008101028.
    1. Houssiau F.A., D’Cruz D., Sangle S., Remy P., Vasconcelos C., Petrovic R., Fiehn C., de Ramon Garrido E., Gilboe I.M., Tektonidou M., et al. Azathioprine versus mycophenolate mofetil for long-term immunosuppression in lupus nephritis: Results from the MAINTAIN Nephritis Trial. Ann. Rheum. Dis. 2010;69:2083–2089. doi: 10.1136/ard.2010.131995.
    1. Tamirou F., D’Cruz D., Sangle S., Remy P., Vasconcelos C., Fiehn C., Ayala Guttierez M., Gilboe I.M., Tektonidou M., Blockmans D., et al. Long-term follow-up of the MAINTAIN Nephritis Trial, comparing azathioprine and mycophenolate mofetil as maintenance therapy of lupus nephritis. Ann. Rheum. Dis. 2016;75:526–531. doi: 10.1136/annrheumdis-2014-206897.
    1. Liu Z., Zhang H., Liu Z., Xing C., Fu P., Ni Z., Chen J., Lin H., Liu F., He Y., et al. Multitarget therapy for induction treatment of lupus nephritis: A randomized trial. Ann. Intern. Med. 2015;162:18–26. doi: 10.7326/M14-1030.
    1. Tamirou F., Lauwerys B.R., Dall’Era M., Mackay M., Rovin B., Cervera R., Houssiau F.A., MAINTAIN Nephritis Trial Investigators A proteinuria cut-off level of 0.7 g/day after 12 months of treatment best predicts long-term renal outcome in lupus nephritis: Data from the MAINTAIN Nephritis Trial. Lupus Sci. Med. 2015;2:e000123. doi: 10.1136/lupus-2015-000123.
    1. Dall’Era M., Cisternas M.G., Smilek D.E., Straub L., Houssiau F.A., Cervera R., Rovin B.H., Mackay M. Predictors of long-term renal outcome in lupus nephritis trials: Lessons learned from the Euro-Lupus Nephritis cohort. Arthritis Rheumatol. 2015;67:1305–1313. doi: 10.1002/art.39026.
    1. Ugolini-Lopes M.R., Seguro L., Castro M., Daffre D., Lopes A.C., Borba E.F., Bonfá E. Early proteinuria response: A valid real-life situation predictor of long-term lupus renal outcome in an ethnically diverse group with severe biopsy-proven nephritis? Lupus Sci. Med. 2017;4:e000213. doi: 10.1136/lupus-2017-000213.
    1. Costedoat-Chalumeau N., Amoura Z., Hulot J.S., Aymard G., Leroux G., Marra D., Lechat P., Piette J.C. Very low blood hydroxychloroquine concentration as an objective marker of poor adherence to treatment of systemic lupus erythematosus. Ann. Rheum. Dis. 2007;66:821–824. doi: 10.1136/ard.2006.067835.
    1. Reyes-Thomas J., Blanco I., Putterman C. Urinary biomarkers in lupus nephritis. Clin. Rev. Allergy Immunol. 2011;40:138–150. doi: 10.1007/s12016-010-8197-z.
    1. Korte E.A., Gaffney P.M., Powell D.W. Contributions of mass spectrometry-based proteomics to defining cellular mechanisms and diagnostic markers for systemic lupus erythematosus. Arthritis Res. Ther. 2012;14:204. doi: 10.1186/ar3701.
    1. Rubinstein T., Pitashny M., Levine B., Schwartz N., Schwartzman J., Weinstein E., Pego-Reigosa J.M., Lu T.Y., Isenberg D., Rahman A., et al. Urinary neutrophil gelatinase-associated lipocalin as a novel biomarker for disease activity in lupus nephritis. Rheumatology. 2010;49:960–971. doi: 10.1093/rheumatology/kep468.
    1. Torres-Salido M.T., Cortés-Hernández J., Vidal X., Pedrosa A., Vilardell-Tarrés M., Ordi-Ros J. Neutrophil gelatinase-associated lipocalin as a biomarker for lupus nephritis. Nephrol. Dial. Transplant. 2014;29:1740–1749. doi: 10.1093/ndt/gfu062.
    1. Rosa R.F., Takei K., Araújo N.C., Loduca S.M., Szajubok J.C., Chahade W.H. Monocyte chemoattractant-1 as a urinary biomarker for the diagnosis of activity of lupus nephritis in Brazilian patients. J. Rheumatol. 2012;39:1948–1954. doi: 10.3899/jrheum.110201.
    1. Schwartz N., Rubinstein T., Burkly L.C., Collins C.E., Blanco I., Su L., Hojaili B., Mackay M., Aranow C., Stohl W., et al. Urinary TWEAK as a biomarker of lupus nephritis: A multicenter cohort study. Arthritis Res. Ther. 2009;11:R143. doi: 10.1186/ar2816.
    1. Somparn P., Hirankarn N., Leelahavanichkul A., Khovidhunkit W., Thongboonkerd V., Avihingsanon Y. Urinary proteomics revealed prostaglandin H(2)D-isomerase, not Zn-α2-glycoprotein, as a biomarker for active lupus nephritis. J. Proteom. 2012;75:3240–3247. doi: 10.1016/j.jprot.2012.03.034.
    1. Gupta R., Yadav A., Misra R., Aggarwal A. Urinary prostaglandin D synthase as biomarker in lupus nephritis: A longitudinal study. Clin. Exp. Rheumatol. 2015;33:694–698.
    1. Vanarsa K., Soomro S., Zhang T., Strachan B., Pedroza C., Nidhi M., Cicalese P., Gidley C., Dasari S., Mohan S., et al. Quantitative planar array screen of 1000 proteins uncovers novel urinary protein biomarkers of lupus nephritis. Ann. Rheum. Dis. 2020;79:1349–1361. doi: 10.1136/annrheumdis-2019-216312.
    1. Alvarado A.S., Malvar A., Lococo B., Alberton V., Toniolo F., Nagaraja H.N., Rovin B.H. The value of repeat kidney biopsy in quiescent Argentinian lupus nephritis patients. Lupus. 2014;23:840–847. doi: 10.1177/0961203313518625.
    1. Alsuwaida A., Husain S., Alghonaim M., AlOudah N., Alwakeel J., Ullah A., Kfoury H. Strategy for second kidney biopsy in patients with lupus nephritis. Nephrol. Dial. Transplant. 2012;27:1472–1478. doi: 10.1093/ndt/gfr517.
    1. Alsuwaida A.O. The clinical significance of serial kidney biopsies in lupus nephritis. Mod. Rheumatol. 2014;24:453–456. doi: 10.3109/14397595.2013.844293.
    1. Zickert A., Sundelin B., Svenungsson E., Gunnarsson I. Role of early repeated renal biopsies in lupus nephritis. Lupus Sci. Med. 2014;1:e000018. doi: 10.1136/lupus-2014-000018.
    1. Parodis I., Adamichou C., Aydin S., Gomez A., Demoulin N., Weinmann-Menke J., Houssiau F.A., Tamirou F. Per-protocol repeat kidney biopsy portends relapse and long-term outcome in incident cases of proliferative lupus nephritis. Rheumatology. 2020;59:3424–3434. doi: 10.1093/rheumatology/keaa129.
    1. Zhang H., Liu Z., Zhou M., Liu Z., Chen J., Xing C., Lin H., Ni Z., Fu P., Liu F., et al. Multitarget Therapy for Maintenance Treatment of Lupus Nephritis. J. Am. Soc. Nephrol. 2017;28:3671–3678. doi: 10.1681/ASN.2017030263.
    1. Rovin B.H., Solomons N., Pendergraft W.F., III, Dooley M.A., Tumlin J., Romero-Diaz J., Lysenko L., Navarra S.V., Huizinga R.B., AURA-LV Study Group A randomized, controlled double-blind study comparing the efficacy and safety of dose-ranging voclosporin with placebo in achieving remission in patients with active lupus nephritis. Kidney Int. 2019;95:219–231. doi: 10.1016/j.kint.2018.08.025.
    1. Arriens C., Polyakova S., Adzerikho I., Randhawa S., Solomons N. OP0277 AURORA phase 3 study demonstrates voclosporin statistical superiority over standard of care in lupus nephritis. Ann. Rheum. Dis. 2020;79:172–173. doi: 10.1136/annrheumdis-2020-eular.5010.
    1. Dooley M.A., Pendergraft W., III, Ginzler E.M., Olsen N.J., Tumlin J., Rovin B.H., Houssiau F.A., Wofsy D., Isenberg D.A., Solomons N., et al. Speed of remission with the use of voclosporin, MMF and low dose steroids: Results of a global lupus nephritis study (abstract) Arthritis Rheumatol. 2016;68(Suppl. 10)
    1. Aurinia FDA Approves Aurinia Pharmaceuticals’ LUPKYNIS (Voclosporin) for Adult Patients with Active Lupus Nephritis [Press Release] [(accessed on 22 January 2021)]; Available online: .
    1. Navarra S.V., Guzmán R.M., Gallacher A.E., Hall S., Levy R.A., Jimenez R.E., Li E.K., Thomas M., Kim H.Y., León M.G., et al. Efficacy and safety of belimumab in patients with active systemic lupus erythematosus: A randomised, placebo-controlled, phase 3 trial. Lancet. 2011;377:721–731. doi: 10.1016/S0140-6736(10)61354-2.
    1. Furie R., Petri M., Zamani O., Cervera R., Wallace D.J., Tegzová D., Sanchez-Guerrero J., Schwarting A., Merrill J.T., Chatham W.W., et al. A phase III, randomized, placebo-controlled study of belimumab, a monoclonal antibody that inhibits B lymphocyte stimulator, in patients with systemic lupus erythematosus. Arthritis Rheum. 2011;63:3918–3930. doi: 10.1002/art.30613.
    1. Dooley M.A., Houssiau F., Aranow C., D’Cruz D.P., Askanase A., Roth D.A., Zhong Z.J., Cooper S., Freimuth W.W., Ginzler E.M., et al. Effect of belimumab treatment on renal outcomes: Results from the phase 3 belimumab clinical trials in patients with SLE. Lupus. 2013;22:63–72. doi: 10.1177/0961203312465781.
    1. Furie R., Rovin B.H., Houssiau F., Malvar A., Teng Y., Contreras G., Amoura Z., Yu X., Mok C.C., Santiago M.B., et al. Two-Year, Randomized, Controlled Trial of Belimumab in Lupus Nephritis. N. Engl. J. Med. 2020;383:1117–1128. doi: 10.1056/NEJMoa2001180.
    1. Sjöwall C., Cöster L. Belimumab may not prevent lupus nephritis in serologically active patients with ongoing non-renal disease activity. Scand. J. Rheumatol. 2014;43:428–430. doi: 10.3109/03009742.2014.887769.
    1. Hui-Yuen J.S., Reddy A., Taylor J., Li X., Eichenfield A.H., Bermudez L.M., Starr A.J., Imundo L.F., Buyon J., Furie R.A., et al. Safety and Efficacy of Belimumab to Treat Systemic Lupus Erythematosus in Academic Clinical Practices. J. Rheumatol. 2015;42:2288–2295. doi: 10.3899/jrheum.150470.
    1. Staveri C., Karokis D., Liossis S.C. New onset of lupus nephritis in two patients with SLE shortly after initiation of treatment with belimumab. Semin. Arthritis Rheum. 2017;46:788–790. doi: 10.1016/j.semarthrit.2016.09.006.
    1. Anjo C., Mascaró J.M., Jr., Espinosa G., Cervera R. Effectiveness and safety of belimumab in patients with systemic lupus erythematosus in a real-world setting. Scand. J. Rheumatol. 2019;48:469–473. doi: 10.1080/03009742.2019.1603324.
    1. Parodis I., Vital E.M., Hassan S.U., Jönsen A., Bengtsson A.A., Eriksson P., Leonard D., Gunnarsson I., Rönnblom L., Sjöwall C. De novo lupus nephritis during treatment with belimumab. Rheumatology. 2020:keaa796. doi: 10.1093/rheumatology/keaa796.
    1. Reddy V., Klein C., Isenberg D.A., Glennie M.J., Cambridge G., Cragg M.S., Leandro M.J. Obinutuzumab induces superior B-cell cytotoxicity to rituximab in rheumatoid arthritis and systemic lupus erythematosus patient samples. Rheumatology. 2017;56:1227–1237. doi: 10.1093/rheumatology/kex067.
    1. Furie R., Aroca G., Alvarez A., Fragoso-Loyo H., Zuta Santillan E., Rovin B., Brunetta P., Schindler T., Hassan I., Cascino M., et al. Two-year results from a randomized; controlled study of obinutuzumab for proliferative lupus nephritis (abstract) Arthritis Rheumatol. 2020;72(Suppl. 10)
    1. Rovin B.H., Furie R., Latinis K., Looney R.J., Fervenza F.C., Sanchez-Guerrero J., Maciuca R., Zhang D., Garg J.P., Brunetta P., et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: The Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012;64:1215–1226. doi: 10.1002/art.34359.
    1. Houssiau F.A. Why will lupus nephritis trials not fail anymore? Rheumatology. 2017;56:677–678. doi: 10.1093/rheumatology/kew252.

Source: PubMed

3
Předplatit