Lupus Nephritis

Back

Practice Essentials

Lupus nephritis is clinically evident in 50-60% of patients with systemic lupus erythematosus (SLE), and it is histologically evident in most SLE patients, even those without clinical manifestations of renal disease. (See the image below.) Evaluating renal function in SLE patients is important because early detection and treatment of renal involvement can significantly improve renal outcome.



View Image

Advanced sclerosis lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class VI (×100, hematoxylin-eosin).

Signs and symptoms

Patients with lupus nephritis may report other symptoms of active SLE (eg, fatigue, fever, rash, arthritis, serositis, or central nervous system [CNS] disease); these are more common with focal proliferative and diffuse proliferative lupus nephritis.

Asymptomatic lupus nephritis 

Active nephritis

Nephritic symptoms related to hypertension and poor renal function (typical of diffuse lupus nephritis):

Nephrotic symptoms related to proteinuria (ypical of membranous lupus nephritis):

Physical findings

See Presentation for more detail.

Diagnosis

Laboratory tests to evaluate renal function in SLE patients include the following:

Laboratory tests for SLE disease activity include the following:

Renal biopsy should be considered in any patient with SLE who has clinical or laboratory evidence of active nephritis, especially upon the first episode of nephritis.

Lupus nephritis is staged according to the classification revised by the International Society of Nephrology (ISN) and the Renal Pathology Society (RPS) in 2003, as follows:

See Workup for more detail.

Management

The principal goal of therapy in lupus nephritis is to normalize renal function or, at least, to prevent the progressive loss of renal function. Therapy differs, depending on the pathologic lesion.

Key points of American College of Rheumatology guidelines for managing lupus nephritis are as follows:

Patients with class V lupus nephritis are generally treated with prednisone for 1-3 months, followed by tapering for 1-2 years if a response occurs. If no response occurs, the drug is discontinued. Immunosuppressive drugs are generally not used unless renal function worsens or a proliferative component is present on renal biopsy samples.

Investigational therapies for lupus nephritis and SLE include the following:

Patients with end-stage renal disease require dialysis and are good candidates for kidney transplantation. Hemodialysis is preferred to peritoneal dialysis.

See Treatment and Medication for more detail.

Background

Lupus nephritis, one of the most serious manifestations of systemic lupus erythematosus (SLE), usually arises within 5 years of diagnosis; however, renal failure rarely occurs before American College of Rheumatology criteria for classification are met.

Lupus nephritis is histologically evident in most patients with SLE, even those without clinical manifestations of renal disease. The symptoms of lupus nephritis are generally related to hypertension, proteinuria, and renal failure. (See Presentation.)

Evaluating renal function in patients with SLE to detect any renal involvement early is important because early detection and treatment can significantly improve renal outcome. Renal biopsy should be considered in any patient with SLE who has clinical or laboratory evidence of active nephritis, especially upon the first episode of nephritis. (See Workup.)

The principal goal of therapy in lupus nephritis is to normalize renal function or, at least, to prevent the progressive loss of renal function. Therapy differs depending on the pathologic lesion. With the advent of more aggressive immunosuppressive and supportive therapy, rates of renal involvement and patient survival are improving. (See Treatment.)

Pathophysiology

Autoimmunity plays a major role in the pathogenesis of lupus nephritis. The immunologic mechanisms include production of autoantibodies directed against nuclear elements. The characteristics of the nephritogenic autoantibodies associated with lupus nephritis are as follows[1] :

These autoantibodies form pathogenic immune complexes intravascularly, which are deposited in glomeruli. Alternatively, autoantibodies may bind to antigens already located in the glomerular basement membrane, forming immune complexes in situ. Immune complexes promote an inflammatory response by activating complement and attracting inflammatory cells, including lymphocytes, macrophages, and neutrophils.[2, 3]

The histologic type of lupus nephritis that develops depends on numerous factors, including the antigen specificity and other properties of the autoantibodies and the type of inflammatory response that is determined by other host factors. In more severe forms of lupus nephritis, proliferation of endothelial, mesangial, and epithelial cells and the production of matrix proteins lead to fibrosis.[4]

Glomerular thrombosis is another mechanism that may play a role in pathogenesis of lupus nephritis, mainly in patients with antiphospholipid antibody syndrome, and is believed to be the result of antibodies directed against negatively charged phospholipid-protein complexes.[2]

Specific strains of a gut commensal may contribute to the immune pathogenesis of lupus nephritis.[5] Azzouz and colleagues analyzed blood and fecal samples from 61 women with SLE and 17 healthy women. On rRNA amplification analysis, patients with SLE displayed a fivefold higher number of Gram-positive fecal bacteria, specifically Ruminococcus gnavus (RG). When the researchers stratified patients with SLE according to organ involvement, those with a history of renal disease demonstrated an abundance of RG-specific amplicon sequence variant compared with those who had no renal impairment.[6]

Etiology

Genetic factors

As with many autoimmune disorders, evidence suggests that genetic predisposition plays an important role in the development of both SLE and lupus nephritis. Multiple genes, many of which are not yet identified, mediate this genetic predisposition (see Table 1 below).[7, 8, 9, 10, 4, 11]

Table 1. Genes Associated With Systemic Lupus Erythematosus



View Table

See Table

SLE is more common in first-degree relatives of patients with SLE (familial prevalence, 10-12%). Concordance rates are higher in monozygotic twins (24-58%) than in dizygotic twins (2-5%), supporting an important role for genetics in the development of SLE. However, the concordance rate in monozygotic twins is not 100%, suggesting that environmental factors trigger development of clinical disease.

Human leukocyte antigen (HLA) class II genes include the following:

Complement genes include the following:

FcγR genes include the following:

Other relevant genes include the following:

Immunologic factors

The initial autoantibody response appears to be directed against the nucleosome, which arises from apoptotic cells.[4, 12, 13]

Patients with SLE have poor clearance mechanisms for cellular debris. Nuclear debris from apoptotic cells induces plasmacytoid dendritic cells to produce interferon-α, which is a potent inducer of the immune system and autoimmunity.[14, 15, 16]

Autoreactive B lymphocytes, which are normally inactive, become active in SLE because of a malfunction of normal homeostatic mechanisms, resulting in escape from tolerance. This leads to the production of autoantibodies. Other autoantibodies, including anti-dsDNA antibodies, develop through a process of epitope spreading. These autoantibodies develop over time, in an orderly fashion, months to years before the onset of clinical SLE.[17]

Epidemiology

Frequency

In a multi-ethnic international cohort of patients enrolled within 15 months (mean, 6 months) after SLE diagnosis and assessed annually, lupus nephritis occurred in 700 of 1827 patients (38.3%). Lupus nephritis was frequently the initial presentation of SLE; it was identified at enrollment in 80.9% of cases. Patients with nephritis were younger, more frequently men and of African, Asian, and Hispanic race/ethnicity.[18]

In a study of a large Spanish registry, lupus nephritis was histologically confirmed in 1092 of 3575 patients with SLE (30.5%). The mean age at lupus nephritis diagnosis was 28.4 years. The risk for lupus nephritis development was significantly higher in men, in younger individuals, and in Hispanics. Patients receiving antimalarials had a significantly lower risk of developing lupus nephritis.[19]

Age-related demographics

Most patients with SLE develop lupus nephritis early in their disease course. SLE is more common among women in the third decade of life, and lupus nephritis typically occurs in patients aged 20-40 years.[20] Children with SLE are at a higher risk of renal disease than adults and tend to sustain more disease damage secondary to more aggressive disease and treatment-associated toxicity.[21, 22, 23]

Sex-related demographics

Because the overall prevalence of SLE is higher in females (ie, female-to-male ratio of 9:1), lupus nephritis is also more common in females; however, clinical renal disease has a worse prognosis and is more common in males with SLE.[20]

Race-related demographics

SLE is more common in African Americans and Asians than in white people, with the highest prevalence in Caribbean people. The prevalence of lupus nephritis is much higher in Asians than in whites with SLE, but the 10-year renal outcome and renal survival rate appear to be better in Asians.[24] Particularly severe lupus nephritis may be more common in African Americans and Asians than in other ethnic groups.[20]

Prognosis

Over the past 4 decades, changes in the treatment of lupus nephritis and general medical care have greatly improved both renal involvement and overall survival. During the 1950s, the 5-year survival rate among patients with lupus nephritis was close to 0%. The subsequent addition of immunosuppressive agents such as intravenous (IV) pulse cyclophosphamide has led to documented 5- and 10-year survival rates as high as 85% and 73%, respectively.[25]

Mortality in patients with end-stage renal disease due to lupus nephritis has declined significantly in recent decades. The mortality rate per 100 patient-years declined from 11.1 in 1995-1999 to 6.7 in 2010-2014. Deaths due to cardiovascular disease declined by 44% and deaths due to infection declined 63%.[26]  

Morbidity associated with lupus nephritis is related to the renal disease itself, as well as to treatment-related complications and comorbidities, including cardiovascular disease and thrombotic events. Progressive renal failure leads to anemia, uremia, and electrolyte and acid-based abnormalities. Hypertension may lead to an increased risk of coronary artery disease and cerebrovascular accident.

Nephrotic syndrome may lead to edema, ascites, and hyperlipidemia, adding to the risk of coronary artery disease and the potential for thrombosis. The findings from one study indicate that patients with lupus nephritis, particularly early-onset lupus nephritis, are at increased risk for morbidity from ischemic heart disease.[27]

In a study of 56 children (< 18 years) with either global or segmental diffuse proliferative lupus nephritis, long-term renal outcomes were similar. Most patients reached adulthood but sustained significant renal damage. Complete remission rates were 50% and 60% in the global and segmental groups, respectively. Renal survival rates, defined as an estimated glomerular filtration rate of ≥60 mL/min/1.73 m2, were 93%, 78%, and 64 % at 1, 5, and 10 years, respectively, and corresponding patient survival rates were 98%, 96%, and 91%, respectively, with similar rates in the global and segmental groups.[28]

Therapy with corticosteroids, cyclophosphamide, and other immunosuppressive agents increases the risk of infection. Long-term corticosteroid therapy may lead to osteoporosis, avascular necrosis, diabetes mellitus, and hypertension, among other complications. Cyclophosphamide therapy may cause cytopenias, hemorrhagic cystitis, infertility, and an increased risk of malignancy.

Biomarkers of renal outcome in lupus nephritis include proteinuria and serum albumin. Studies have shown that a proteinuria cut-off of less than 0.7 or 0.8 g/day at 12 months predicts good long-term renal outcome. Domingues et al reported that serum albumin > 3.7 g/dL at 12 months predicts favorable renal outcome at 48 months.[29]

 

History

Patients with active lupus nephritis often have other symptoms of active systemic lupus erythematosus (SLE), including fatigue, fever, rash, arthritis, serositis, or central nervous system (CNS) disease. These are more common with focal proliferative and diffuse proliferative lupus nephritis.[30]

Some patients have asymptomatic lupus nephritis; however, during regular follow-up, laboratory abnormalities such as elevated serum creatinine levels, low albumin levels, or urinary protein or sediment suggests active lupus nephritis. This is more typical of mesangial or membranous lupus nephritis.

Symptoms related to active nephritis may include peripheral edema secondary to hypertension or hypoalbuminemia. Extreme peripheral edema is more common in persons with diffuse or membranous lupus nephritis, as these renal lesions are commonly associated with heavy proteinuria.[25]

Other symptoms directly related to hypertension that are commonly associated with diffuse lupus nephritis include headache, dizziness, visual disturbances, and signs of cardiac decompensation.

Physical Examination

With focal and diffuse lupus nephritis, the physical examination may reveal evidence of generalized active SLE with the presence of a rash, oral or nasal ulcers, synovitis, or serositis. Signs of active nephritis are also common.

With active lupus nephritis, patients have hypertension, peripheral edema, and, occasionally, cardiac decompensation.

With membranous lupus nephritis, signs of an isolated nephrotic syndrome are common. These include peripheral edema, ascites, and pleural and pericardial effusions without hypertension.

Approach Considerations

Evaluating renal function in patients with systemic lupus erythematosus (SLE) to detect any renal involvement early is important because early detection and treatment can significantly improve renal outcome.[25]

Renal biopsy should be considered in any patient with SLE who has clinical or laboratory evidence of active nephritis, especially upon the first episode of nephritis.[25, 31]

Lupus nephritis is staged according to the classification revised by the International Society of Nephrology (ISN) and the Renal Pathology Society (RPS) in 2003. This classification is based on light microscopy, immunofluorescence, and electron microscopy findings from renal biopsy specimens.

Laboratory Tests for Renal Function in SLE

Laboratory tests to evaluate renal function in SLE patients include the following:

In an international study, Smith and colleagues reported that a panel of novel urinary biomarkers can accurately identify active lupus nephritis in children.[32] These authors concluded that the optimal biomarker panel would include the following:

Laboratory Tests for SLE Disease Activity

SLE disease activity can be evaluated by assessing antibodies to double-stranded DNA (dsDNA), complement (C3, C4, and CH50), erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP) levels.

The CRP level is generally not elevated in patients with SLE, even with active disease, unless the patient has significant arthritis or infection.[25] Generally, elevated ESR and anti-dsDNA and depressed C3 and C4 levels are associated with active nephritis, especially focal and diffuse lupus nephritis. Clinically relevant lupus nephritis is associated with a 30% decrease in creatinine clearance, proteinuria of greater than 1000 mg/d, and renal biopsy findings indicating active lupus nephritis.

Anti-nucleosome antibodies appear early in the course of the autoimmune response in SLE, they have high sensitivity and specificity for a diagnosis of SLE, and the titers correlate with disease activity.[33, 34, 35] Anti-C1q antibodies are associated with lupus nephritis; higher titers correlate with active renal disease.[36, 37]

Anti-C1q antibodies have a sensitivity of 44-100% and a specificity of 70-92% in active renal disease (SLE); in combination with low C3 and C4 levels, these may be the predictors of renal flares in patients with SLE.[38] Although anti-DNA antibodies were more sensitive than anti-C1q antibodies for active lupus nephritis (75% vs 53%, respectively), anti-C1q antibodies were more specific (84% vs 49%, respectively); the negative predictive value of negative anti-DNA and anti-C1q antibodies for active lupus nephritis was 91%.[39]

Renal Biopsy

Renal biopsy may be useful in patients with recurrent episodes of nephritis, depending on the clinical circumstances. By revealing the histologic pattern and stage of disease (activity and chronicity), renal biopsy is useful in determining prognosis and treatment. Findings from a thorough clinical and laboratory evaluation can be used to predict the histologic type of lupus nephritis in approximately 70-80% of patients; however, this is not accurate enough, in view of the toxicity of some of the treatment protocols.

A good rule is to perform a renal biopsy if the findings will potentially alter patient management. If a particular patient has other manifestations of SLE (eg, severe central nervous system [CNS] or hematologic involvement) and will be treated with cyclophosphamide, biopsy may not be necessary but should still be considered because it may help predict renal outcome.

Sampling error can occur during a renal biopsy. Thus, the results of the biopsy should always be evaluated for consistency with the clinical and laboratory presentation of the patient.

The experience of pathologists in reading lupus nephritis biopsy specimens varies considerably. Studies have suggested that the most consistent readers are in larger medical centers with substantial populations of patients with SLE.

Staging

The classification of lupus nephritis was revised by the International Society of Neurology and the Renal Pathology Society (ISN/RPS) in 2003 and is based on light microscopy, immunofluorescence, and electron microscopy findings from renal biopsy specimens. The ISN/RPS classification itself is based on earlier classifications by the World Health Organization (WHO) in 1974 and 1982 (see Table 2 below).[40]

Table 2. International Society of Nephrology/Renal Pathology Society 2003 Classification of Lupus Nephritis



View Table

See Table

In addition to the pathologic classification, activity and chronicity indices are scored pathologically and predict the renal prognosis—that is, the progression of renal disease (see Table 3 below). The activity index reflects the state of active inflammation observed at biopsy, which may be reversible with medical therapy. The chronicity index reflects the amount of fibrosis and scarring, which are unlikely to respond to therapy. Renal lesions with a high activity index are more likely to respond to aggressive therapy, whereas renal lesions with high chronicity are not.

These indices in the table below serve as a prognostic tool and a general guide to therapy. Signs of activity justify aggressive medical therapy because such therapy may arrest or reverse the pathologic changes. Signs of chronicity suggest irreversibility, and aggressive therapy is less likely to affect the outcome. The activity and chronicity indices are evaluated at a single point in time, and renal lesions may transform from one class to another either spontaneously or as a result of treatment.

Table 3. Active and Chronic Glomerular Lesions



View Table

See Table

See the images below.



View Image

Mesangial proliferative lupus nephritis with moderate mesangial hypercellularity. International Society of Nephrology/Renal Pathology Society 2003 cla....



View Image

Focal lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class III (×100, hematoxylin-eosin).



View Image

Focal lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class III (×200, immunofluorescence).



View Image

Diffuse lupus nephritis with hypertensive vascular changes. International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxyl....



View Image

Diffuse lupus nephritis with early crescent formation. International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eo....



View Image

Diffuse lupus nephritis with extensive crescent formation (rapidly progressive glomerulonephritis). International Society of Nephrology/Renal Patholog....



View Image

Membranous lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class V (×200, hematoxylin-eosin).



View Image

Membranous lupus nephritis showing thickened glomerular basement membrane. International Society of Nephrology/Renal Pathology Society 2003 class V (×....



View Image

Advanced sclerosis lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class VI (×100, hematoxylin-eosin).

Approach Considerations

The principal goal of therapy in lupus nephritis is to normalize renal function or, at least, to prevent the progressive loss of renal function. Therapy differs depending on the pathologic lesion.[25, 41] It is important to treat extrarenal manifestations and other variables that may affect the kidneys.

Corticosteroid therapy should be instituted if the patient has clinically significant renal disease. Use immunosuppressive agents, particularly cyclophosphamide, azathioprine, or mycophenolate mofetil, if the patient has aggressive proliferative renal lesions, as they improve the renal outcome. Immunosuppressives can also be used if the patient has an inadequate response or excessive sensitivity to corticosteroids.[41, 42, 43]

Calcineurin inhibitors, especially tacrolimus, have demonstrated benefit in lupus nephritis. However, most studies have been limited to Asian patients, and further research is required on long-term benefits and disadvantages.[44, 45, 46]

Treat hypertension aggressively. On the basis of beneficial effects in other nephropathies, angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs) have been routinely used to treat proteinuria in lupus nephritis.

Alter the diet according to the presence of hypertension, hyperlipidemia, and renal insufficiency. Restrict fat intake or use lipid-lowering therapy such as statins for hyperlipidemia secondary to nephrotic syndrome. Restrict protein intake if renal function is significantly impaired.

Administer calcium supplementation to prevent osteoporosis if the patient is on long-term corticosteroid therapy, and consider adding a bisphosphonate (depending on renal function).

Avoid drugs that affect renal function, including nonsteroidal anti-inflammatory drugs (NSAIDs), especially in patients with elevated creatinine levels. Nonacetylated salicylates can be used to safely treat inflammatory symptoms in patients with renal disease.

Patients with active lupus nephritis should avoid pregnancy, because it may worsen their renal disease and because certain medications used in the treatment may be teratogenic.[47] In women who desire pregnancy, the following approach is advised[48] :

Patients with end-stage renal disease (ESRD), sclerosis, and a high chronicity index based on renal biopsy findings are unlikely to respond to aggressive therapy. In these cases, focus therapy on extrarenal manifestations of systemic lupus erythematosus (SLE) and on possible kidney transplantation.

Pharmacotherapy for Lupus Nephritis Based on Stage

Classes I and II

Minimal mesangial (class I) lupus nephritis requires no specific therapy.[25]

Mesangial proliferative (class II) lupus nephritis may require treatment if proteinuria is greater than 1000 mg/day. Consider prednisone in low-to-moderate doses (ie, 20-40 mg/day) for 1-3 months, with subsequent taper.

Classes III and IV

Patients with either focal (class III) or diffuse (class IV) lupus nephritis are at high risk of progressing to ESRD and thus require aggressive therapy.

Administer prednisone 1 mg/kg/day for at least 4 weeks, depending on clinical response. Then, taper it gradually to a daily maintenance dose of 5-10 mg/day for approximately 2 years. In acutely ill patients, intravenous (IV) methylprednisolone at a dosage of up to 1000 mg/day for 3 days may be used to initiate corticosteroid therapy.

In patients who do not respond to corticosteroids alone, who have unacceptable toxicity to corticosteroids, who have worsening renal function, who have severe proliferative lesions, or who have evidence of sclerosis on renal biopsy specimens, use immunosuppressive drugs in addition to corticosteroids.

Both cyclophosphamide and azathioprine are effective in proliferative lupus nephritis, although cyclophosphamide is apparently more effective in preventing progression to ESRD. Mycophenolate mofetil has been shown to be at least as effective as IV cyclophosphamide, with less toxicity, in patients with focal or diffuse lupus nephritis who have stable renal function.[49, 50] It may be used alone[49, 50] or sequentially after a 6-month course of IV cyclophosphamide.[51]

Appel et al studied 370 patients with lupus nephritis in a randomized open-label study and found no significant difference in clinical improvement was observed with mycophenolate mofetil compared with IV cyclophosphamide.[52] The study included induction and maintenance therapy, and both study groups received prednisone.

Administer IV cyclophosphamide monthly for 6 months and every 2-3 months thereafter, depending on clinical response. The usual duration of therapy is 2-2.5 years. Reduce the dose if the creatinine clearance is less than 30 mL/min. Adjust the dose depending on the hematologic response.[53, 54] The gonadotropin-releasing hormone analogue leuprolide acetate has been shown to protect against ovarian failure.[55]

Azathioprine can also be used as a second-line agent, with dose adjustments depending on hematologic response.

Mycophenolate mofetil was found to be superior to azathioprine in maintaining control and preventing relapses of lupus nephritis in patients who have responded to induction therapy.[56]

In a 10-year follow-up of the MAINTAIN Nephritis Trial, which compared azathioprine and mycophenolate mofetil as maintenance therapy of proliferative lupus nephritis, Tamirou  and colleagues found that the two treatments resulted in similar outcomes. Two deaths and one case of end-stage renal disease developed in the azathioprine group, versus three deaths and three cases of  end-stage renal disease in the mycophenolate mofetil group.[57]

Class V

Patients with membranous lupus nephritis are generally treated with prednisone for 1-3 months, followed by tapering for 1-2 years if a response occurs. If no response occurs, the drug is discontinued. Immunosuppressive drugs are generally not used unless renal function worsens or a proliferative component is present on renal biopsy samples. Some clinical evidence indicates that azathioprine, cyclophosphamide, cyclosporine, and chlorambucil are effective in reducing proteinuria. Mycophenolate mofetil may also be effective.

In a study of membranous lupus nephritis, 38 patients were treated with corticosteroids and azathioprine; after 12 months of treatment, 67% of patients had a complete remission and 22% had a partial remission, with only 11% resistant to treatment.[58] Long-term follow-up of 12 years showed 19 episodes of renal flares. Retreatment with corticosteroids and azathioprine showed similar responses.

Investigational Therapies for Lupus Nephritis and SLE

Rituximab

Rituximab, a B-lymphocyte–depleting therapy, appears to be effective in SLE and is being investigated as a treatment for SLE and lupus nephritis. Several small case series of rituximab have shown benefit in SLE and lupus nephritis.[59, 60, 61, 62]

More recently, however, a randomized, double-blind, phase II/III trial of rituximab in moderately-to-severely active SLE (EXPLORER) failed to show differences compared with placebo, although a beneficial effect of rituximab was noted in the African-American and Hispanic subgroups.[63]   A randomized, double-blind, phase III trial of rituximab in active proliferative lupus nephritis (LUNAR) showed that rituximab therapy resulted in more responders and greater reductions in anti-DNA antibodies in increases in C3 and C4 levels, but it did not improve clinical outcomes after 1 year of treatment.[64]

Other anti-CD20 monoclonal antibodies

Other anti-CD20 monoclonal antibodies have been used experimentally for lupus nephritis; for example, in patients who respond to rituximab but develop intolerable adverse effects. These include the following[65] :

Belimumab

Belimumab (Benlysta) is an anti–B-lymphocyte stimulator [BLyS] monoclonal antibody).[67] It has been found to have beneficial effects on clinical and laboratory parameters in patients with active SLE.[68] In addition, the number of B cells and serum IgM were reduced over time.[69]

Belimumab was approved by the US Food and Drug Administration (FDA) for use in patients with active SLE who are autoantibody-positive and are receiving standard therapy, including corticosteroids, antimalarials, immunosuppressives, and NSAIDs.

Voclosporin

In a phase II study, an induction regimen that combined the novel calcineurin inhibitor voclosporin with mycophenolate mofetil and low-dose oral corticosteroids led to complete remission in almost a third of patients with acute lupus nephritis, and to partial remission in the majority.[70] A phase III trial of voclosporin is in progress.[71]

Atacicept

Atacicept is a TACI-Ig fusion protein that inhibits BLyS and a proliferation-inducing ligand [APRIL]).[67] In early phase studies, atacicept was demonstrated to have biologic effects in patients with SLE, resulting in a dose-dependent reduction in B cells and immunoglobulin levels.[72]

Abetimus

Abetimus is a B-lymphocyte tolerogen that was found to be ineffective in preventing flares of lupus nephritis in a large controlled trial, although it did reduce levels of anti-DNA antibodies.[73]

Anticytokine therapies

Various anticytokine therapies have been proposed, including monoclonal antibodies directed against the following[67] :

Management of End-Stage Renal Disease

Patients with ESRD require dialysis and are good candidates for kidney transplantation (see Renal Transplantation). Patients with ESRD secondary to SLE represent 1.5% of all patients on dialysis in the United States. The survival rate among patients on dialysis is fair (5-year survival rate, 60-70%) and is comparable with that among patients on dialysis who do not have SLE.

Hemodialysis is preferred to peritoneal dialysis; several studies have documented higher levels of antibodies to double-stranded DNS (dsDNA), more thrombocytopenia, and higher steroid requirements in patients with SLE and ESRD who are on peritoneal dialysis. Hemodialysis also has anti-inflammatory effects with decreased T-helper lymphocyte levels. SLE is generally quiescent in patients on hemodialysis, although flares, including rash, arthritis, serositis, fever, and leukopenia may occur, necessitating specific treatment.

Renal Transplantation

Patients with SLE account for 3% of all renal transplantations in the United States. It is important ensure that the patient does not have active SLE disease at the time of transplantation. A 3-month period of dialysis is usually prudent in the event of spontaneous renal recovery.

Substantial evidence shows that patients with SLE fare worse than patients without SLE in terms of graft survival. Living-related allografts show better outcomes than cadaveric allografts. In patients with SLE, reasons for a more severe outcome after transplantation include recurrent lupus nephritis and concomitant antiphospholipid antibody syndrome resulting in allograft loss.[74]

Consultations

It is frequently advisable to consult a nephrologist for renal biopsy or, if desired, for help in the management of renal disease.

The experience of pathologists in reading lupus nephritis biopsy specimens varies considerably. The most consistent readers of these specimens are usually found in larger academic centers that have substantial populations of patients with SLE.

Guidelines Summary

Guidelines for managing lupus nephritis have been issued by the American College of Rheumatology.[75]  Key points of the guidelines are as follows:

Joint guidelines for the management of adult and pediatric lupus nephritis have also been issued by European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA). The EULAR/ERA-EDTA recommendations include the following[77] :

A European initiative, the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) project, has published guidelines on the diagnosis and treatment of childhood-onset lupus nephritis.[78, 79]

Diagnostic recommendations from SHARE include the following[79] :

SHARE recommends complete renal response as the treatment goal in lupus nephritis, with an early-morning urinary protein/creatinine ratio of less than 50 mg/mmol and normal renal function. Patients should achieve a partial response within 6 to 12 months after starting treatment.[79]  Treatment recommendations include the following:

Recommendations for management based on the International Society of Nephrology/Renal Pathology Society 2003 classification system include the following[79] :

Medication Summary

Corticosteroids are used in all patients with clinically significant renal disease. Immunosuppressive agents, particularly cyclophosphamide, azathioprine, and mycophenolate mofetil, are used in patients with aggressive renal lesions because they improve the renal outcome. They may also be used in patients with inadequate response or excessive toxicity to corticosteroids. Cyclosporine has been used in some cases.

Prednisone

Clinical Context:  Prednisone is commonly used to treat inflammatory manifestations of SLE. Treatment of clinically significant lupus nephritis should include moderate-to-high doses initially.

Methylprednisolone (Medrol, Solu-Medrol, A-Methapred)

Clinical Context:  Methylprednisolone is used in much the same manner as prednisone, but it has less mineralocorticoid effects and should be considered in patients with edema. The parenteral IV dosage form is used in the inpatient setting.

Class Summary

Corticosteroids are very useful in controlling acute inflammatory manifestations of systemic lupus erythematosus (SLE). Alone, they may be adequate in treating milder forms of lupus nephritis with a lower risk of progressive renal dysfunction, such as minimal mesangial lupus nephritis, mesangial proliferative lupus nephritis, early focal lupus nephritis, or membranous lupus nephritis. Oral corticosteroids can be used in most patients. If adequate absorption is a concern (eg, bowel edema in a patient with nephrosis), intravenous (IV) methylprednisolone can be used.

Cyclophosphamide

Clinical Context:  Cyclophosphamide is indicated for treatment of most patients with focal lupus nephritis or diffuse lupus nephritis. Although it has significant toxicity, it has been shown to prevent the progression of nephritis and improve renal outcome.

Azathioprine (Imuran, Azasan)

Clinical Context:  Azathioprine is useful in moderate-to-severe lupus nephritis. It improves renal outcome, but it does not appear to be as effective as cyclophosphamide, although it is less toxic.

Mycophenolate mofetil (CellCept) or mycophenolic acid (Myfortic)

Clinical Context:  Mycophenolate mofetil is an option for induction therapy with class II/IV lupus nephritis. It has generally been well tolerated and, in several studies, has been as effective as (and possibly more effective than) more traditional therapies, including cyclophosphamide and azathioprine, with less toxicity. The American College of Rheumatology guidelines recommend mycophenolate mofetil as the preferred agent for African Americans and Hispanics.

Hydroxychloroquine (Plaquenil)

Clinical Context:  The exact anti-inflammatory mechanism of action of hydroxychloroquine is not well understood. It is thought to elicit anti-inflammatory effects in vivo by antagonizing histamine and serotonin and inhibits prostaglandin synthesis. In vitro studies suggest hydroxychloroquine may inhibit chemotaxis of PMN leukocytes, macrophages, and eosinophils. The American College of Rheumatology guidelines recommend that all patients with SLE and nephritis be treated with a background of hydroxychloroquine, unless contraindicated.

Class Summary

In particular, cyclophosphamide, mycophenolate, and azathioprine are used in patients with aggressive renal lesions (eg, focal or diffuse lupus nephritis) because they improve renal outcome. These agents can also be used in patients with inadequate response or excessive toxicity to corticosteroids. Mycophenolate mofetil has been shown to be effective for treatment of lupus nephritis. Mycophenolate mofetil was found to be superior to azathioprine in maintaining control and preventing relapses of lupus nephritis in patients who have responded to induction therapy.[56]

What is lupus nephritis?What are the signs and symptoms of lupus nephritis?What is the presentation of asymptomatic lupus nephritis?What is the presentation of active lupus nephritis?What are the physical findings suggestive of lupus nephritis?Which lab tests are performed in the evaluation of lupus nephritis?When is renal biopsy indicated in the evaluation of lupus nephritis?How is lupus nephritis staged?What is the principal goal of therapy in lupus nephritis?What are the American College of Rheumatology treatment guidelines for lupus nephritis?How is class V lupus nephritis treated?What are investigational therapies for lupus nephritis?What is the role of dialysis in the management of lupus nephritis?What is lupus nephritis?What is the principal goal of therapy in lupus nephritis?What is the role of autoimmunity in the pathogenesis of lupus nephritis?Which factors affect the development of histologic type of lupus nephritis?What is the role of glomerular thrombosis in the pathogenesis of lupus nephritis?Which Fc?R genes play a role in the etiology of lupus nephritis?What is the role of genetics in the etiology of lupus nephritis?What is the role of human leukocyte antigen (HLA) class II genes in the etiology of lupus nephritis?Which complement genes have an etiologic role in lupus nephritis?What genes play a role in the etiology of lupus nephritis?What is the role of immunologic factors in the etiology of lupus nephritis?What is the prevalence of lupus nephritis?How does the prevalence of lupus nephritis vary by age?How does the incidence of lupus nephritis vary by sex?What are the racial predilections for lupus nephritis?Why has the prognosis of lupus nephritis improved over the years?What is the morbidity associated with lupus nephritis?What is the prognosis of nephrotic syndrome in lupus nephritis?What is the prognosis of global or segmental diffuse proliferative lupus nephritis?Which therapeutic agents increase the risk of infection in lupus nephritis?What is the clinical history characteristic of lupus nephritis?Which physical findings are characteristic of lupus nephritis?What are diagnostic considerations for lupus nephritis?Which conditions should be included in the differential diagnosis of lupus nephritis?What are the differential diagnoses for Lupus Nephritis?How is lupus nephritis staged?Which lab tests are used to evaluate renal function in systemic lupus erythematosus (SLE) with suspected lupus nephritis?What is the optimal biomarker panel in the assessment of active lupus nephritis in children?How is systemic lupus erythematosus (SLE) disease activity assessed in lupus nephritis?What is the role of renal biopsy in the workup of lupus nephritis?What is the ISN/RPS classification of lupus nephritis?What is the principal goal of therapy in lupus nephritis?What is the role of corticosteroid therapy for lupus nephritis?What is the role of calcineurin inhibitors in the treatment of lupus nephritis?How is hypertension managed in patients with lupus nephritis?What is the role of calcium supplementation in the treatment of lupus nephritis?Which drugs should be avoided by patients with lupus nephritis?What is the approach to treatment of active lupus nephritis in women planning pregnancy?Which patients with lupus nephritis are unlikely to respond to aggressive therapy?What are the American College of Rheumatology guidelines for managing lupus nephritis?What are the European League Against Rheumatism and European Renal Association-European Dialysis and Transplant Association (EULAR/ERA-EDTA treatment guidelines for lupus nephritis?What are the treatment options for classes I and II lupus nephritis?What are the treatment options for classes III and IV lupus nephritis?What is the role of cyclophosphamide in the treatment of lupus nephritis?What is the efficacy of cyclophosphamide in the treatment of lupus nephritis?What are the treatment options for class V lupus nephritis?Which anti-CD20 monoclonal antibodies are used in the treatment of lupus nephritis?What is the role of rituximab in the treatment of lupus nephritis?What is the role of belimumab (Benlysta) in the treatment of lupus nephritis?What is the role of voclosporin in the treatment of lupus nephritis?What is the role of atacicept in the treatment of lupus nephritis?What is the role of abetimus in the treatment of lupus nephritis?What is the role of anticytokine therapies in the treatment of lupus nephritis?How is end-stage renal disease (ESRD) managed in lupus nephritis?When is renal transplantation indicated in the treatment of lupus nephritis?Which specialist consultations are beneficial for the treatment of lupus nephritis?Which organization has published guidelines for the diagnosis and treatment of childhood-onset lupus nephritis?What are the diagnostic recommendations from the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) for lupus nephritis?What are the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) guidelines recommended goals for treatment of lupus nephritis?What are the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) treatment guidelines for lupus nephritis?Which medications are used in the treatment of lupus nephritis?Which medications in the drug class Immunosuppressives are used in the treatment of Lupus Nephritis?Which medications in the drug class Corticosteroids are used in the treatment of Lupus Nephritis?

Author

Lawrence H Brent, MD, Associate Professor of Medicine, Sidney Kimmel Medical College of Thomas Jefferson University; Chair, Program Director, Department of Medicine, Division of Rheumatology, Albert Einstein Medical Center

Disclosure: Stock ownership for: Johnson & Johnson.

Coauthor(s)

Arati S Karhadkar, MD, Consulting Physician, Rheumatology Associates, Ltd

Disclosure: Nothing to disclose.

Eric Bloom, MD, Division Chief of Nephrology, Nephrology Fellowship Program Director, Attending Physician, Division of Nephrology, Department of Internal Medicine, Albert Einstein Medical Center

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Ajay K Singh, MB, MRCP, MBA, Associate Professor of Medicine, Harvard Medical School; Director of Dialysis, Renal Division, Brigham and Women's Hospital; Director, Brigham/Falkner Dialysis Unit, Faulkner Hospital

Disclosure: Nothing to disclose.

Chief Editor

Vecihi Batuman, MD, FASN, Huberwald Professor of Medicine, Section of Nephrology-Hypertension, Tulane University School of Medicine; Chief, Renal Section, Southeast Louisiana Veterans Health Care System

Disclosure: Nothing to disclose.

Additional Contributors

Carlos J Lozada, MD, Director of Rheumatology Fellowship Training Program, Professor of Clinical Medicine, Department of Medicine, Division of Rheumatology and Immunology, University of Miami, Leonard M Miller School of Medicine

Disclosure: Received honoraria from Pfizer for consulting; Received grant/research funds from AbbVie for other; Received honoraria from Heel for consulting.

Acknowledgements

The authors and editors of Medscape Reference gratefully acknowledge the contributions of previous author Irene Viola, MD, to the development and writing of the source article.

References

  1. Yung S, Chan TM. Anti-DNA antibodies in the pathogenesis of lupus nephritis--the emerging mechanisms. Autoimmun Rev. 2008 Feb. 7(4):317-21. [View Abstract]
  2. Davidson A, Berthier C, Kretzler M. Pathogenetic mechanisms in lupus nephritis. Wallace DJ, Hahn BH, eds. Dubois' Lupus Erythematosus and Related Syndromes. 8th ed. Philadelphia, PA: Elsevier Saunders; 2013. 237-55.
  3. Grande JP. Mechanisms of progression of renal damage in lupus nephritis: pathogenesis of renal scarring. Lupus. 1998. 7(9):604-10. [View Abstract]
  4. Lisnevskaia L, Murphy G, Isenberg D. Systemic lupus erythematosus. Lancet. 2014 Nov 22. 384 (9957):1878-1888. [View Abstract]
  5. Garcia J. Gut Microbes May Predict Lupus Flares. Medscape Medical News. February 26, 2019. Available at https://www.medscape.com/viewarticle/909553
  6. Azzouz D, Omarbekova A, Heguy A, Schwudke D, Gisch N, Rovin BH, et al. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Ann Rheum Dis. 2019 Feb 19. [View Abstract]
  7. Mohan C, Putterman C. Genetics and pathogenesis of systemic lupus erythematosus and lupus nephritis. Nat Rev Nephrol. 2015 Jun. 11 (6):329-41. [View Abstract]
  8. Harley JB, Kelly JA, Kaufman KM. Unraveling the genetics of systemic lupus erythematosus. Springer Semin Immunopathol. 2006 Oct. 28(2):119-30. [View Abstract]
  9. Harley JB, Alarcón-Riquelme ME, Criswell LA, Jacob CO, Kimberly RP, Moser KL, et al. Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet. 2008 Feb. 40(2):204-10. [View Abstract]
  10. Crow MK. Collaboration, genetic associations, and lupus erythematosus. N Engl J Med. 2008 Feb 28. 358(9):956-61. [View Abstract]
  11. Kaiser R, Criswell LA. Genetics research in systemic lupus erythematosus for clinicians: methodology, progress, and controversies. Curr Opin Rheumatol. 2010 Mar. 22(2):119-25. [View Abstract]
  12. Mohan C, Adams S, Stanik V, Datta SK. Nucleosome: a major immunogen for pathogenic autoantibody-inducing T cells of lupus. J Exp Med. 1993 May 1. 177(5):1367-81. [View Abstract]
  13. Muller S, Dieker J, Tincani A, Meroni PL. Pathogenic anti-nucleosome antibodies. Lupus. 2008. 17(5):431-6. [View Abstract]
  14. Crow MK. Interferon pathway activation in systemic lupus erythematosus. Curr Rheumatol Rep. 2005 Dec. 7(6):463-8. [View Abstract]
  15. Rönnblom L, Eloranta ML, Alm GV. The type I interferon system in systemic lupus erythematosus. Arthritis Rheum. 2006 Feb. 54(2):408-20. [View Abstract]
  16. Rönnblom L, Pascual V. The innate immune system in SLE: type I interferons and dendritic cells. Lupus. 2008. 17(5):394-9. [View Abstract]
  17. Arbuckle MR, McClain MT, Rubertone MV, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003 Oct 16. 349(16):1526-33. [View Abstract]
  18. Hanly JG, O'Keeffe AG, Su L, Urowitz MB, Romero-Diaz J, et al. The frequency and outcome of lupus nephritis: results from an international inception cohort study. Rheumatology (Oxford). 2016 Feb. 55 (2):252-62. [View Abstract]
  19. Galindo-Izquierdo M, Rodriguez-Almaraz E, Pego-Reigosa JM, et al. Characterization of Patients With Lupus Nephritis Included in a Large Cohort From the Spanish Society of Rheumatology Registry of Patients With Systemic Lupus Erythematosus (RELESSER). Medicine (Baltimore). 2016 Mar. 95 (9):e2891. [View Abstract]
  20. Lim SS, Drenkard C. The Epidemiology of Lupus. Wallace DJ, Hahn BH, eds. Dubois' Lupus Erythematosus and Related Syndromes. 8th ed. Philadelphia, PA: Elsevier Saunders; 2013. 8-24.
  21. Gloor JM. Lupus nephritis in children. Lupus. 1998. 7(9):639-43. [View Abstract]
  22. Brunner HI, Gladman DD, Ibañez D, Urowitz MD, Silverman ED. Difference in disease features between childhood-onset and adult-onset systemic lupus erythematosus. Arthritis Rheum. 2008 Feb. 58(2):556-62. [View Abstract]
  23. Bogdanovic R, Nikolic V, Pasic S, Dimitrijevic J, Lipkovska-Markovic J, Eric-Marinkovic J. Lupus nephritis in childhood: a review of 53 patients followed at a single center. Pediatr Nephrol. 2004 Jan. 19(1):36-44. [View Abstract]
  24. Yap DY, Chan TM. Lupus Nephritis in Asia: Clinical Features and Management. Kidney Dis (Basel). 2015 Sep. 1 (2):100-9. [View Abstract]
  25. Dooley MA. Clinical and epidemiologic features of lupus nephritis. Wallace DJ, Hahn BH, eds. Dubois' Lupus Erythematosus and Related Syndromes. 8th ed. Philadelphia, PA: Elsevier Saunders; 2013. 438-54.
  26. Jorge A, Wallace ZS, Zhang Y, Lu N, Costenbader KH, Choi HK. All-Cause and Cause-Specific Mortality Trends of End-Stage Renal Disease due to Lupus Nephritis from 1995 to 2014. Arthritis Rheumatol. 2018 Sep 18. [View Abstract]
  27. Faurschou M, Mellemkjaer L, Starklint H, et al. High risk of ischemic heart disease in patients with lupus nephritis. J Rheumatol. 2011 Nov. 38(11):2400-5. [View Abstract]
  28. Rianthavorn P, Buddhasri A. Long-term renal outcomes of childhood-onset global and segmental diffuse proliferative lupus nephritis. Pediatr Nephrol. 2015 Nov. 30 (11):1969-76. [View Abstract]
  29. Domingues V, Levinson BA, Bornkamp N, Goldberg JD, Buyon J, Belmont HM. Serum albumin at 1 year predicts long-term renal outcome in lupus nephritis. Lupus Sci Med. 2018. 5 (1):e000271. [View Abstract]
  30. Pisetsky DS, Gilkeson G, St. Clair EW. Systemic lupus erythematosus. Diagnosis and treatment. Med Clin North Am. 1997 Jan. 81(1):113-28. [View Abstract]
  31. Grande JP, Balow JE. Renal biopsy in lupus nephritis. Lupus. 1998. 7(9):611-7. [View Abstract]
  32. Smith EM, Jorgensen AL, Midgley A, Oni L, Goilav B, Putterman C, et al. International validation of a urinary biomarker panel for identification of active lupus nephritis in children. Pediatr Nephrol. 2016 Sep 3. [View Abstract]
  33. Simón JA, Cabiedes J, Ortiz E, Alcocer-Varela J, Sánchez-Guerrero J. Anti-nucleosome antibodies in patients with systemic lupus erythematosus of recent onset. Potential utility as a diagnostic tool and disease activity marker. Rheumatology (Oxford). 2004 Feb. 43(2):220-4. [View Abstract]
  34. Su Y, Jia RL, Han L, Li ZG. Role of anti-nucleosome antibody in the diagnosis of systemic lupus erythematosus. Clin Immunol. 2007 Jan. 122(1):115-20. [View Abstract]
  35. Bigler C, Lopez-Trascasa M, Potlukova E, Moll S, Danner D, Schaller M, et al. Antinucleosome antibodies as a marker of active proliferative lupus nephritis. Am J Kidney Dis. 2008 Apr. 51(4):624-9. [View Abstract]
  36. Marto N, Bertolaccini ML, Calabuig E, Hughes GR, Khamashta MA. Anti-C1q antibodies in nephritis: correlation between titres and renal disease activity and positive predictive value in systemic lupus erythematosus. Ann Rheum Dis. 2005 Mar. 64(3):444-8. [View Abstract]
  37. Sinico RA, Radice A, Ikehata M, Giammarresi G, Corace C, Arrigo G, et al. Anti-C1q autoantibodies in lupus nephritis: prevalence and clinical significance. Ann N Y Acad Sci. 2005 Jun. 1050:193-200. [View Abstract]
  38. Sinico RA, Rimoldi L, Radice A, Bianchi L, Gallelli B, Moroni G. Anti-C1q autoantibodies in lupus nephritis. Ann N Y Acad Sci. 2009 Sep. 1173:47-51. [View Abstract]
  39. Mok CC, Ho LY, Leung HW, Wong LG. Performance of anti-C1q, antinucleosome, and anti-dsDNA antibodies for detecting concurrent disease activity of systemic lupus erythematosus. Transl Res. 2010 Dec. 156(6):320-5. [View Abstract]
  40. Weening JJ, D'Agati VD, Schwartz MM, Seshan SV, Alpers CE, Appel GB. The classification of glomerulonephritis in systemic lupus erythematosus revisited. J Am Soc Nephrol. 2004 Feb. 15(2):241-50. [View Abstract]
  41. Houssiau FA, Ginzler EM. Current treatment of lupus nephritis. Lupus. 2008. 17(5):426-30. [View Abstract]
  42. Dooley MA, Falk RJ. Immunosuppressive therapy of lupus nephritis. Lupus. 1998. 7(9):630-4. [View Abstract]
  43. Dooley MA, Ginzler EM. Newer therapeutic approaches for systemic lupus erythematosus: immunosuppressive agents. Rheum Dis Clin North Am. 2006 Feb. 32(1):91-102, ix. [View Abstract]
  44. Kronbichler A, Neumann I, Mayer G. Moderator's view: the use of calcineurin inhibitors in the treatment of lupus nephritis. Nephrol Dial Transplant. 2016 Sep 1. [View Abstract]
  45. Mok CC, Jayne D. Pro: The use of calcineurin inhibitors in the treatment of lupus nephritis. Nephrol Dial Transplant. 2016 Sep 1. [View Abstract]
  46. Fernandez Nieto M, Jayne DR, Mok CC. Con: The use of calcineurin inhibitors in the treatment of lupus nephritis. Nephrol Dial Transplant. 2016 Sep 1. [View Abstract]
  47. Lightstone L, Hladunewich MA. Lupus Nephritis and Pregnancy: Concerns and Management. Semin Nephrol. 2017 Jul. 37 (4):347-353. [View Abstract]
  48. Moroni G, Doria A, Giglio E, Tani C, Zen M, Strigini F, et al. Fetal outcome and recommendations of pregnancies in lupus nephritis in the 21st century. A prospective multicenter study. J Autoimmun. 2016 Aug 2. [View Abstract]
  49. Chan TM, Li FK, Tang CS, et al. Efficacy of mycophenolate mofetil in patients with diffuse proliferative lupus nephritis. Hong Kong-Guangzhou Nephrology Study Group. N Engl J Med. 2000 Oct 19. 343(16):1156-62. [View Abstract]
  50. Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill JT. Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. N Engl J Med. 2005 Nov 24. 353(21):2219-28. [View Abstract]
  51. Contreras G, Pardo V, Leclercq B, et al. Sequential therapies for proliferative lupus nephritis. N Engl J Med. 2004 Mar 4. 350(10):971-80. [View Abstract]
  52. Appel GB, Contreras G, Dooley MA, et al. Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol. 2009 May. 20(5):1103-12. [View Abstract]
  53. Gourley MF, Austin HA 3rd, Scott D, et al. Methylprednisolone and cyclophosphamide, alone or in combination, in patients with lupus nephritis. A randomized, controlled trial. Ann Intern Med. 1996 Oct 1. 125(7):549-57. [View Abstract]
  54. Ciruelo E, de la Cruz J, Lopez I, Gomez-Reino JJ. Cumulative rate of relapse of lupus nephritis after successful treatment with cyclophosphamide. Arthritis Rheum. 1996 Dec. 39(12):2028-34. [View Abstract]
  55. Somers EC, Marder W, Christman GM, Ognenovski V, McCune WJ. Use of a gonadotropin-releasing hormone analog for protection against premature ovarian failure during cyclophosphamide therapy in women with severe lupus. Arthritis Rheum. 2005 Sep. 52(9):2761-7. [View Abstract]
  56. Dooley MA, Jayne D, Ginzler EM, et al. Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis. N Engl J Med. 2011 Nov 17. 365(20):1886-95. [View Abstract]
  57. Tamirou F, D'Cruz D, Sangle S, Remy P, Vasconcelos C, Fiehn C, 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. 2015 Mar 10. [View Abstract]
  58. Mok CC, Ying KY, Yim CW, Ng WL, Wong WS. Very long-term outcome of pure lupus membranous nephropathy treated with glucocorticoid and azathioprine. Lupus. 2009 Oct. 18(12):1091-5. [View Abstract]
  59. Silverman GJ. Anti-CD20 therapy in systemic lupus erythematosus: a step closer to the clinic. Arthritis Rheum. 2005 Feb. 52(2):371-7. [View Abstract]
  60. Looney RJ, Anolik JH, Campbell D, Felgar RE, Young F, Arend LJ. B cell depletion as a novel treatment for systemic lupus erythematosus: a phase I/II dose-escalation trial of rituximab. Arthritis Rheum. 2004 Aug. 50(8):2580-9. [View Abstract]
  61. Leandro MJ, Cambridge G, Edwards JC, Ehrenstein MR, Isenberg DA. B-cell depletion in the treatment of patients with systemic lupus erythematosus: a longitudinal analysis of 24 patients. Rheumatology (Oxford). 2005 Dec. 44(12):1542-5. [View Abstract]
  62. Ng KP, Leandro MJ, Edwards JC, Ehrenstein MR, Cambridge G, Isenberg DA. Repeated B cell depletion in treatment of refractory systemic lupus erythematosus. Ann Rheum Dis. 2006 Jul. 65(7):942-5. [View Abstract]
  63. Merrill JT, Neuwelt CM, Wallace DJ, et al. Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial. Arthritis Rheum. 2010 Jan. 62(1):222-33. [View Abstract]
  64. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum. 2012 Apr. 64 (4):1215-26. [View Abstract]
  65. Looney RJ, Anolik J, Sanz I. New therapies for systemic lupus erythematosus: cellular targets. Rheum Dis Clin North Am. 2006 Feb. 32(1):201-15, xi. [View Abstract]
  66. Haarhaus ML, Svenungsson E, Gunnarsson I. Ofatumumab treatment in lupus nephritis patients. Clin Kidney J. 2016 Aug. 9 (4):552-5. [View Abstract]
  67. Kirou KA, Salmon JE, Crow MK. Soluble mediators as therapeutic targets in systemic lupus erythematosus: cytokines, immunoglobulin receptors, and the complement system. Rheum Dis Clin North Am. 2006 Feb. 32(1):103-19, ix. [View Abstract]
  68. Wallace DJ, Stohl W, Furie RA, et al. A phase II, randomized, double-blind, placebo-controlled, dose-ranging study of belimumab in patients with active systemic lupus erythematosus. Arthritis Rheum. 2009 Sep 15. 61(9):1168-78. [View Abstract]
  69. Jacobi AM, Huang W, Wang T, et al. Effect of long-term belimumab treatment on B cells in systemic lupus erythematosus: extension of a phase II, double-blind, placebo-controlled, dose-ranging study. Arthritis Rheum. 2010 Jan. 62(1):201-10. [View Abstract]
  70. Rovin BH, Solomons N, Pendergraft WF 3rd, Dooley MA, Tumlin J, Romero-Diaz J, 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. 2018 Nov 5. [View Abstract]
  71. Aurinia Renal Response in Active Lupus With Voclosporin (AURORA). ClinicalTrials.gov. Available at https://clinicaltrials.gov/ct2/show/NCT03021499. October 25, 2018; Accessed: December 23, 2018.
  72. Dall'Era M, Chakravarty E, Wallace D, et al. Reduced B lymphocyte and immunoglobulin levels after atacicept treatment in patients with systemic lupus erythematosus: results of a multicenter, phase Ib, double-blind, placebo-controlled, dose-escalating trial. Arthritis Rheum. 2007 Dec. 56(12):4142-50. [View Abstract]
  73. Furie R. Abetimus sodium (riquent) for the prevention of nephritic flares in patients with systemic lupus erythematosus. Rheum Dis Clin North Am. 2006 Feb. 32(1):149-56, x. [View Abstract]
  74. Gołębiewska J, Dębska-Ślizień A, Bułło-Piontecka B, Rutkowski B. Outcomes in Renal Transplant Recipients With Lupus Nephritis-A Single-Center Experience and Review of the Literature. Transplant Proc. 2016 Jun. 48 (5):1489-93. [View Abstract]
  75. [Guideline] Hahn BH, McMahon MA, Wilkinson A, Wallace WD, Daikh DI, Fitzgerald JD, et al. American college of rheumatology guidelines for screening, treatment, and management of lupus nephritis. Arthritis Care Res (Hoboken). 2012 Jun. 64(6):797-808. [View Abstract]
  76. Tsakonas E, Joseph L, Esdaile JM, Choquette D, Senécal JL, Cividino A, et al. A long-term study of hydroxychloroquine withdrawal on exacerbations in systemic lupus erythematosus. The Canadian Hydroxychloroquine Study Group. Lupus. 1998. 7(2):80-5. [View Abstract]
  77. [Guideline] 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 paediatric lupus nephritis. Ann Rheum Dis. 2012 Nov. 71 (11):1771-82. [View Abstract]
  78. Reuters. New Guidelines Focus on Diagnosing, Treating Lupus Nephritis in Kids. Medscape Medical News. Available at https://www.medscape.com/viewarticle/885734. September 17, 2017; Accessed: November 16, 2017.
  79. Groot N, de Graeff N, Marks SD, Brogan P, Avcin T, Bader-Meunier B, et al. European evidence-based recommendations for the diagnosis and treatment of childhood-onset lupus nephritis: the SHARE initiative. Ann Rheum Dis. 2017 Dec. 76 (12):1965-1973. [View Abstract]
  80. Ward MM. Recent clinical trials in lupus nephritis. Rheum Dis Clin North Am. 2014 Aug. 40 (3):519-35, ix. [View Abstract]
  81. Witter FR. Management of the high-risk lupus pregnant patient. Rheum Dis Clin North Am. May 2007. 33(2):253-65, v-vi. [View Abstract]
  82. Condon MB, Ashby D, Pepper RJ, Cook HT, Levy JB, Griffith M, 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 Aug. 72(8):1280-6. [View Abstract]

Advanced sclerosis lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class VI (×100, hematoxylin-eosin).

Mesangial proliferative lupus nephritis with moderate mesangial hypercellularity. International Society of Nephrology/Renal Pathology Society 2003 class II (×200, hematoxylin-eosin).

Focal lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class III (×100, hematoxylin-eosin).

Focal lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class III (×200, immunofluorescence).

Diffuse lupus nephritis with hypertensive vascular changes. International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eosin).

Diffuse lupus nephritis with early crescent formation. International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eosin).

Diffuse lupus nephritis with extensive crescent formation (rapidly progressive glomerulonephritis). International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eosin).

Membranous lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class V (×200, hematoxylin-eosin).

Membranous lupus nephritis showing thickened glomerular basement membrane. International Society of Nephrology/Renal Pathology Society 2003 class V (×200, silver stain).

Advanced sclerosis lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class VI (×100, hematoxylin-eosin).

Mesangial proliferative lupus nephritis with moderate mesangial hypercellularity. International Society of Nephrology/Renal Pathology Society 2003 class II (×200, hematoxylin-eosin).

Focal lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class III (×100, hematoxylin-eosin).

Focal lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class III (×200, immunofluorescence).

Diffuse lupus nephritis with hypertensive vascular changes. International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eosin).

Diffuse lupus nephritis with early crescent formation. International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eosin).

Diffuse lupus nephritis with extensive crescent formation (rapidly progressive glomerulonephritis). International Society of Nephrology/Renal Pathology Society 2003 class IV (×200, hematoxylin-eosin).

Membranous lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class V (×200, hematoxylin-eosin).

Membranous lupus nephritis showing thickened glomerular basement membrane. International Society of Nephrology/Renal Pathology Society 2003 class V (×200, silver stain).

Advanced sclerosis lupus nephritis. International Society of Nephrology/Renal Pathology Society 2003 class VI (×100, hematoxylin-eosin).

Gene Locus Gene Name Gene Product
1p13.2 PTPN22 Lymphoid-specific protein tyrosine phosphatase
1q21-q23 CRP CRP
1q23 FCGR2A, FCGR2B FcγRIIA (R131), FcγRIIB
1q23 FCGR3A, FCGR3B FcγRIIIA (V176), FcγRIIIB
1q31-q32 IL10 IL-10
1q36.12 C1QB C1q deficiency
2q32.2-q32.3 STAT4 Signal transducer and activator of transcription 4
2q33 CTLA4 Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4)
6p21.3 HLA-DRB1 HLA-DRB1: DR2/*1501, DR3/*0301C1q deficiency
6p21.3 C2, C4A, C4B C2, C4 deficiencies
6p21.3 TNF TNF-a (promoter, -308)
10q11.2-q21 MBL2 Mannose-binding lectin
CRP = C-reactive protein; HLA = human leukocyte antigen; IL = interleukin; TNF = tumor necrosis factor.
Class I



Minimal mesangial lupus nephritis



Light microscopy findingsNormal
Immunofluorescence electron microscopy findingsMesangial immune deposits
Clinical manifestationsMild proteinuria
Class II



Mesangial proliferative lupus nephritis



Light microscopy findingsPurely mesangial hypercellularity or mesangial matrix expansion with mesangial immune deposits
Immunofluorescence electron microscopy findingsMesangial immune deposits; few immune deposits in subepithelial or subendothelial deposits possible
Clinical manifestationsMild renal disease such as asymptomatic hematuria or proteinuria that usually does not warrant specific therapy
Class III



Focal lupus nephritis



Class III (A)



Active lesions - Focal proliferative lupus nephritis



Class III (A/C)



Active and chronic lesions - Focal proliferative and sclerosing lupus nephritis



Class III (C)



Chronic inactive lesions - Focal sclerosing lupus nephritis



Light microscopy findingsActive or inactive focal, segmental, or global glomerulonephritis involving < 50% of all glomeruli
Immunofluorescence electron microscopy findingsSubendothelial and mesangial immune deposits
Clinical manifestationsActive generalized SLE and mild-to-moderate renal disease with hematuria and moderate proteinuria in many patients; worsening renal function in significant minority, potentially progressing to class IV lupus nephritis
Class IV



Diffuse lupus nephritis



Class IV-S (A)



Active lesions - Diffuse segmental proliferative lupus nephritis



Class IV-G (A)



Active lesions - Diffuse global proliferative lupus nephritis



Class IV-S (A/C)



Active and chronic lesions - Diffuse segmental proliferative and sclerosing lupus nephritis



Class IV-G (A/C)



Active and chronic lesions - Diffuse global proliferative and sclerosing lupus nephritis



Class IV-S (C)



Chronic inactive lesions with scars - Diffuse segmental sclerosing lupus nephritis



Class IV-G (C)



Chronic inactive lesions with scars - Diffuse global sclerosing lupus nephritis



Light microscopy findingsActive or inactive diffuse, segmental or global glomerulonephritis involving = 50% of all glomeruli; subdivided into diffuse segmental (class IV-S) when = 50% of involved glomeruli have segmental lesions (involving less than half of glomerular tuft) and diffuse global (class IV-G) when = 50% of involved glomeruli have global lesions
Immunofluorescence electron microscopy findingsSubendothelial immune deposits
Clinical manifestationsClinical evidence of renal disease including hypertension, edema, active urinary sediment, worsening renal function, and nephrotic range proteinuria in most cases; active extrarenal SLE in many patients
Class V



Membranous lupus nephritis



Light microscopy findingsDiffuse thickening of glomerular basement membrane without inflammatory infiltrate; possibly, subepithelial deposits and surrounding basement membrane spikes on special stains, including silver and trichrome; may occur in combination with class II or IV; may show advanced sclerosis
Immunofluorescence electron microscopy findingsSubepithelial and intramembranous immune deposits; subendothelial deposits present only when associated proliferative component is present
Clinical manifestationsClinical and laboratory features of nephrotic syndrome, usually without manifestations of active SLE
Class VI



Advanced sclerosis lupus nephritis



Light microscopy findingsAdvanced glomerular sclerosis involving = 90% of glomeruli, interstitial fibrosis, and tubular atrophy, all morphological manifestations of irreversible renal injury
Clinical manifestationsSignificant renal insufficiency or end-stage renal disease in most cases; unlikely to respond to medical therapy
SLE = systemic lupus erythematosus.
Activity Index Chronicity Index
• Endocapillary hypercellularity with or without leukocyte infiltration; luminal reduction



• Karyorrhexis



• Fibrinoid necrosis



• Rupture of glomerular basement membrane



• Cellular or fibrocellular crescents



• Subendothelial deposits on light microscopy



• Intraluminal immune aggregates



• Glomerular sclerosis; segmental, global



• Fibrous adhesion



• Fibrous crescents