Aktuelle Rheumatologie 2020; 45(02): 150-162
DOI: 10.1055/a-1117-2920
Übersichtsarbeit
© Georg Thieme Verlag KG Stuttgart · New York

Nierenbeteiligung bei Kollagenosen – Teil 1: Lupus-Nephritis

Renal Involvement in Connective Tissue Diseases – part 1: Lupus Nephritis
Peter Oelzner
1   Klinik für Innere Medizin III, Universitätsklinikum Jena
,
Kerstin Amann
2   Abteilung Nephropathologie, Universitätsklinikum Erlangen
,
Gunter Wolf
3   Nephrologie, Klinik für Innere Medizin III, Universitätsklinikum Jena
› Author Affiliations
Further Information

Publication History

Publication Date:
02 April 2020 (online)

Zusammenfassung

Die Lupus-Nephritis (LN) tritt in Abhängigkeit von Ethnizität und Geschlecht in bis zu 50% der Patienten mit systemischem Lupus erythematodes (SLE) auf und ist die prognostisch entscheidende Organmanifestation bei SLE. Pathogenetisch wichtige Aspekte des SLE sind eine multifaktoriell bedingte Störung der Clearance von im Rahmen von Apoptose und NETose anfallendem Autoantigen, was in der Entwicklung einer Autoimmunreaktion resultiert, eine Amplifizierung der pathologischen Immunreaktion durch eine Überaktivierung des Typ-I-Interferon-Signalweges und eine Zytokinimbalance. An der Niere manifestiert sich der pathogenetische Prozess in Form einer Immunkomplexglomerulonephritis. Entscheidend für die Prognose der LN sind frühzeitige Diagnose und umgehende Therapieeinleitung. Die Auswahl der medikamentösen Therapie basiert grundsätzlich auf dem Befund der Nierenbiopsie. Bei Vorliegen einer proliferativen LN (Klasse III und IV, auch in Kombination mit einer membranösen LN) erfolgt eine Remissionsinduktion mit einer intravenösen low-dose Cyclophosphamid (CYC) – Therapie (6 x 500 mg) oder mit Mycophenolatmofetil (MMF) kombiniert mit initial hoch dosierten Glukokortikoiden (GK), gefolgt von einer Remissionserhaltung mit Azathioprin oder MMF. Bei Therapie-refraktärer Situation erfolgt der Wechsel von CYC auf MMF oder umgekehrt. Alternativ können auch Rituximab oder Calcineurin-Inhibitoren eingesetzt werden. Bei anderen Formen der LN wird das therapeutische Vorgehen wesentlich durch die Entwicklung der Nierenfunktion und das Ausmaß der Proteinurie bestimmt. Zusätzlich kommen supportive Massnahmen, wie der generelle Einsatz von Hydroxychloroquin, ACE-Hemmer oder Angiotensin-Rezeptor-Blocker in Abhängigkeit vom Ausmaß der Proteinurie und vom Blutdruck, sowie Maßnahmen zur Thromboembolie-, Osteoporose- und Infektionsprophylaxe zur Anwendung.

Ziele der Therapie sind eine möglichst komplette renale Remission, die Vermeidung chronischer Schäden und eine effektive Reduktion von GK. Eine komplette Remission, welche sich über den Erhalt einer normalen Nierenfunktion und eine effektive Reduktion der Proteinurie definiert, wird in ca. 50–60% erreicht. Dies unterstreicht einerseits die Effektivität der aktuellen Therapie, beleuchtet aber auch die Notwendigkeit neuer Therapiestrategien, gerade auch in Anbetracht der hohen Rate chronischer Schäden.

Neue therapeutische Ansätze wie Multitarget-Therapie und neue Protokolle zur B-Zell-Depletion und -Neutralisation sowie weitgehend GK-freie Behandlungsprinzipien zielen auf eine noch effektivere und Nebenwirkungs-ärmere Therapie der LN.

Abstract

Lupus nephritis (LN) occurs dependent on ethnicity and gender in up to 50% of patients with systemic lupus erythematosus (SLE) and represents the prognostic most important organ manifestation in SLE. Pathogenetic important aspects of SLE are a multifactorial caused disturbance of the clearance of autoantigen released in the context of apoptosis and NETosis resulting in the development of autoimmune reaction, an amplification of the pathologic immune reaction by hyperactivation of type I-interferon signaling and cytokine imbalance. The pathogenetic process in the kidney results in immune complex glomerulonephritis.

Early diagnosis and initiation of therapy are of critical importance for the prognosis of LN. The selection of therapy is based principally on the findings in renal biopsy. In the case of proliferative LN (class III or IV, also in combination with membranous LN) a remission induction with low-dose cyclophosphamide (CYC)-pulse therapy (6 x 500 mg) or with mycophenolatofetil (MMF) in combination with initial high dose glucocorticoids (GC) is followed by a maintenance therapy with azathioprine or MMF. In refractory situations a switch from CYC to MMF or conversely is indicated. Alternatively rituximab or calcineurin inhibitors can also be used. In other forms of LN the therapeutic procedure is essentially determinated by the course of renal function and the extent of proteinuria. In addition supportive procedures such as the general use of hydroxychloroquine, ACE inhibitors or angiotensin rezeptor blockers in dependence of the extent of proteinuria and of blood pressure and prophylactic measures to prevent thromboembolism, osteoporosis and infections come into use. Aims of therapy are a preferably complete renal remission, prevention of chronic damage and an effective reduction of GC dose. A complete remission which is defined by the preservation of normal renal function and a substantial reduction of proteinuria is achieved in approximately 50–60%. This underlines the effectiveness of the curent therapy on one hand but illuminates the need of new therapeutic strategies on the other, especially in view of the high rate of damage.

New therapeutic approaches such multitarget therapy, new protocolls of B cell depletion and B cell neutralization and GC free treatment principles aim at a more effective and side effect poor therapy of LN.

 
  • Literatur

  • 1 Lewis MJ, Jawad AS. The SLE review series: working for a better standard of care. The effect of ethnicity and genetic ancestry on the epidemiology, clinical features and outcome of systemic lupus erythematosus. Rheumatology 2017; 56: i67-i77
  • 2 Dörner T, Furie R. Novel paradigms in systemic lupus erythematosus. Lancet 2019; 393: 2344-2358
  • 3 Hanly JG, OʼKeeffe AG, Su L. et al. The frequency and outcome of lupus nephritis. Results from an international inception cohort study. Rheumatology 2016; 55: 252-262
  • 4 Lorenz G, Desai J, Anders HJ. Lupus Nephritis: update on mechanisms of systemic autoimmunity and kidney immunopathology. Curr Opin Nephrol Hypertens 2014; 23: 211-217
  • 5 Janivani A, Ashrfa-Ganjouei A, Aslani S. et al. Exploring the etiopathogenesis of systemic lupus erythematosus: a genetic perspective. Immunogenetics 2019; 71: 283-297
  • 6 Finzel S, Schaffer S, Rizzi M. et al. Pathogenese des systemischen Lupus erythematodes. Z Rheumatol 2018; 77: 789-798
  • 7 Tenbrock K. Pathogenese und neue Therapieansätze beim systemischen Lupus erythematodes. Z Rheumatol 2014; 73: 890-896
  • 8 Jung Y, Suh CH. Incomplete clearance of apoptotic cells in systemic lupus erythematosus. Pathogenetic role and potential biomarker. Int J Rheumatol 2015; 18: 294-303
  • 9 Lu Q, Lemke G. Homeostatic regulation of the immune system by receptor tyrosine kinasis of the Tyro 3 family. Science 2001; 293: 306-311
  • 10 Tsokos GC, Lo MS, Costa Reis P. et al. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2016; 12: 716-730
  • 11 Mistry P, Kaplan MJ. Cell Death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin Immunol 2017; 185: 59-73
  • 12 Thorlacius GE, Wahren-Herlenius M, Ronnblom L. An update on the role of type I interferons in systemic lupus erythematosus and Sjogrenʼs syndrome. Curr Opin Rheumatol 2018; 30: 471-481
  • 13 Steri M, Orru V, Idda ML. et al. Overexpression oft the cytokine BAFF and autoimmunity risk. N Engl J Med 2017; 376: 1615-1626
  • 14 Weening JJ, DʼAgati VD, Schwartz MM. et al. International Soiety of Nephrology Working Group on the Classification of Lupus Nephritis; Renal Pathology Society Working Group on the Classification of Lupus Nephritis. The classification of glomerulonephritis in systemic lupus erythematosus revisited. Kidney Int 2004; 65: 521-530
  • 15 Bajema IM, Wilhelmus S, Alpers CE. et al. Revision oft 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
  • 16 Aringer M, Costenbader K, Daikh D. et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Ann Rheum Dis 2019; 78: 1151-1159
  • 17 Franklyn K, Lau CS, Navarra SV. et al. Definition and initial validation of a Lupus Low Disease Activity State (LLDAS). Ann Rheum Dis 2016; 75: 1615-1621
  • 18 Wilhelm TR, Magder LS, Petri M. Remission in systemic lupus erythematosus: durable remission is rare. Ann Rheum Dis 2017; 76: 547-553
  • 19 Gordon C, Jayne D, Pusey C. et al. European consensus statement on the terminology used in the management of lupus glomerulonephritis. Lupus 2009; 18: 257-263
  • 20 Bertsias GK, Tektonidou M, Amoura Z. et al. Joint European League against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA) recommendations for the management of adult and pediatric lupus nephritis. Ann Rheum Dis 2012; 71: 1171-1782
  • 21 Hahn BH, McMahon MA, Wilkinson A. et al. American College of Rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res 2012; 64: 797-808
  • 22 Bertsias G, Boumpas DT. Update on the management of lupus nephritis: let the treatment fit the patient. Nat Clin Pract Rheumatol 2008; 4: 464-472
  • 23 Fanouriakis A, Kostopoulou M, Alunno A. et al. 2019 update of the EULAR recommendations for the management of systemic lupus erythematosus. Ann Rheum Dis 2019; 78: 736-745
  • 24 Houssiau FA, Vasconcelos C, DʼCruz D. 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
  • 25 Houssiau FA, Vasconcelos C, DʼCruz D. 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
  • 26 Ginzler EM, Dooley MA, Aranow C. et al. Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl J Med 2005; 353: 2219-2228
  • 27 Appel GB, Contreras G, Dooley MA. et al. and the Aspreva Lupus Management Study Group Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol 2009; 20: 1103-1112
  • 28 Rathi M, Goyal A, Jaryal A. et al. Comparison of low-dose intravenous cyclophosphamide with oral mycophenolate mofetil in the treatment of lupus nephritis. Kidney Int 2016; 89: 235-242
  • 29 Tamirou F, DʼCruz D, Sangle S. et al. and the MAINTAIN Nephritis Trial Group. 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
  • 30 Dooley MA, Jayen D, Ginzler EM. et al. ALMS Group Mycophenolate versus azathioprine as maintemance therapy for lupus nephritis. N Engl J Med 2011; 365: 1886-1895
  • 31 Maneiro JR, Lopez-Canoa N, Salgado E. et al. Maintenance therapy of lupus nephritis with mycophenolate or azathioprine: systematic review and meta-analysis. Rheumatology 2014; 53: 834-838
  • 32 Deng J, Xie H, Zhu L. et al. Maintenance therapy for lupus nephritis with mycophenolate mofetil or azathioprine. A meta-analysis. Clin Nephrol 2019; 91: 172-179
  • 33 Mok CC, Ying KY, Yim CW. et al. Tacrolimus versus mycophenolate mofetil for induction therapy of lupus nephritis: a randomized controlled trial and long-term follow-up. Ann Rheum Dis 2016; 75: 30-36
  • 34 Liu Z, Zhang H, Liu Z. et al. Multitarget therapy for induction treatment of lupus nephritis. A randomized trial. Ann Intern Med 2015; 162: 18-26
  • 35 Zhang H. Liu Zhou M. et al. Multitarget therapy for maintenance treatment of lupus nephritis. J Am Soc Nephrol 2017; 28: 3671-3678
  • 36 Park DJ, Kang JH, Lee KE. et al. Efficacy and safety of mycophenolate mofetil and tacrolimus combination therapy in patients with lupus nephritis: a nationwide multicentre study. Clin Exp Rheumatol 2019; 37: 89-96
  • 37 Rovin BH, Solomons N, Pendergraft WF. et al. 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
  • 38 Rovin BH, Furie R, Latinis K. et al. LUNAR Investigator Group 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
  • 39 Mysler EF, Spindler AJ, Guzman R. et al. Efficacy and safety of ocrelizumab in active proliferative lupus nephritis: results from a randomized, double-blind, phase III study. Arthritis Rheum 2013; 65: 2368-2379
  • 40 Almaani S, Rovin BH. B cell therapy in lupus nephritis: an overview. Nephrol Dial Transplant 2019; 34: 22-29
  • 41 Condon MB, Ashby D, Pepper RJ. et al. Prospective observational single-centre cohort study to evaluate the effectiveness of treating lupus nephritis with rituximab and mycophenolate mofetil but no oral steroids. Ann Rheum Dis 2013; 72: 1280-1286
  • 42 Weidenbusch M, Römmele C, Schröttle A. et al. Beyond the LUNAR trial. Efficacy of rituximab in refractory lupus nephritis. Nephrol Dial Transplant 2013; 28: 106-111
  • 43 Iwata S, Saito K, Hirata S. et al. Efficacy and safety of anti-CD20 antibody rituximab for patients with refractory systemic lupus erythematosus. Lupus 2018; 27: 802-811
  • 44 Iaccarino L, Bartoloni E, Carli L. et al. Efficacy and safety of off-label use of rituximab in refractory lupus: data from the Italian Multicentre Registry. Clin Exp Rheumatol 2015; 33: 449-456
  • 45 Dooley MA, Houssiau F, Aranow C. et al. Effect of belimumab treatment on renal oucomes: results from the phase 3 belimumab clinical trials in patients with SLE. Lupus 2013; 22: 63-72
  • 46 Ginzler EM, Wax S, Rajeswaran A. et al. Atacicept in combination with MMF and corticosteroid in lupus nephritis. Results of a prematurely terminated trial. Arthritis Res Ther. 2012 doi: 10.1186/ar3738
  • 47 Furie RA, Leon G, Thomas M. et al. A phase 2, randomized, placebo-controlled clinical trial of blisibimod, an inhibitor of B cell activating factor, in patients with moderat-to-severe systemic lupus erythematosus, the PEARL-SC study. Ann Rheum Dis 2015; 74: 1667-1675
  • 48 Furie R, Niccholls K, Cheng TT. et al. Efficacy and safety of abatacept in lupus nephritis: a twelve-month, randomized, double-blind study. Arthritis Rheumatol 2014; 66: 379-389
  • 49 Askanase AD, Byron M, Keyes-Elstein LL. et al. ACCESS Trial Group Treatment of lupus nephritis with abatacept: the Abatacept and Cyclophosphamide Combination Efficacy and Safety Study. Arthritis Rheumatol 2014; 66: 3096-3104
  • 50 Davidson A, Aranow C, Mackay M. Lupus nephritis: challenges and progress. Curr Opin Rheumatol 2019; 31: 682-688
  • 51 Morand EF, Furie R, Tanaka Y. et al. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med. 2019 doi: 10.1056/NEJMoa1912196
  • 52 Van Vollenhoven RF, Hahn BH, Tsokos GC. et al. Maintenance of efficacy and safety of ustekinumab through 1 year in a randomized phase II trial of patients with active systemic lupus erythematosus. Arthritis Rheumatol. 2019 doi: 10.1002/art.41179