You are on page 1of 13

Clin Exp Nephrol (2016) 20:1–13

DOI 10.1007/s10157-015-1179-y

REVIEW ARTICLE

Lupus nephritis: an update


Tasnim F. Imran1 • Frederick Yick1 • Suneet Verma1,2 • Christopher Estiverne1 •

Chinonye Ogbonnaya-Odor1 • Srikanth Thiruvarudsothy1 • Alluru S. Reddi1,2 •


Neil Kothari1

Received: 15 April 2015 / Accepted: 2 October 2015 / Published online: 16 October 2015
Ó Japanese Society of Nephrology 2015

Abstract Lupus nephritis (LN) is an inflammatory con- including pathogenesis, classification, and clinical mani-
dition of the kidneys that encompasses various patterns of festations. We will focus, though, on discussion of the
renal disease including glomerular and tubulointerstitial established as well as emerging therapies for patients with
pathology. It is a major predictor of poor prognosis in proliferative and membranous lupus nephritis.
patients with systemic lupus erythematosus (SLE). Genetic
factors, including several predisposing loci, and environ- Keywords Lupus nephritis  Systemic lupus
mental factors, such as EBV and ultraviolet light, have erythematosus  Renal lupus
been implicated in the pathogenesis. It carries a high
morbidity and mortality if left untreated. Renal biopsy
findings are utilized to guide treatment. Optimizing risk Introduction
factors such as proteinuria and hypertension with renin-
angiotensin receptor blockade is crucial. Immunosuppres- Lupus nephritis (LN) refers to inflammation of the kidney
sive therapy is recommended for patients with focal or that encompasses diverse patterns of renal disease includ-
diffuse proliferative lupus nephritis (Class III or IV) dis- ing glomerular, tubulointerstitial and vascular pathology
ease, and certain patients with membranous LN (Class V) [1]. Renal involvement in systemic lupus erythematosus
disease. Over the past decade, immunosuppressive thera- (SLE) may be present in approximately 60 % of adults,
pies have significantly improved long-term outcomes, but with 25–50 % of patients presenting with clinical renal
the optimal therapy for LN remains to be elucidated. disease at the time of diagnosis [2]. Most patients affected
Cyclophosphamide-based regimens, given concomitantly are female, and younger than 50 years of age. However,
with corticosteroids, have improved survival significantly. male patients tend to have more frequent renal involvement
Even though many patients achieve remission, the risk of and greater severity of disease [3, 4]. The prevalence of
relapse remains considerably high. Other treatments SLE ranges from 1.4 to 21.9 %; incidence is estimated to
include hydroxychloroquine, mycofenolate mofetil, and be 7.4–159.4 cases per 100,000 person-years. The inci-
biologic therapies such as Belimumab, Rituximab, and dence varies depending on the studied population. The
Abatacept. In this paper, we provide a review of LN, cumulative incidence of LN is higher in people of Asian
(55 %), African (51 %), and Hispanic (43 %) ancestry as
compared with Caucasians (14 %) [5].
& Tasnim F. Imran With the advent of advanced therapies, the 5-year renal
imranta@njms.rutgers.edu survival rate of LN patients has shown continuous
1
improvement from 44 % in 1953–1969, 82 % in
Department of Medicine, New Jersey Medical School,
1990–1995, to nearly 96 % today [6, 7]. Lupus nephritis,
Rutgers, The State University of New Jersey, 185 South
Orange Avenue, Newark, NJ 07103, USA when occurring early in the course of SLE, is considered a
2 major predictor of poor prognosis [8]. In about 5 % of the
Division of Nephrology, New Jersey Medical School,
Rutgers, The State University of New Jersey, 185 South cases, LN may appear more than 5 years after the onset of
Orange Avenue, Newark, NJ 07103, USA SLE (delayed LN). Delayed LN has a higher association

123
2 Clin Exp Nephrol (2016) 20:1–13

with Sjogren syndrome (SS), lung involvement, and Evidence suggests that nucleosomes from apoptotic
antiphospholipid syndrome as compared with early LN [9]. cells largely contribute to the immunogenic material tar-
Although the etiology remains unclear, genetic, hormonal geted by autoantibodies [15]. Initially, apoptosis was
and environmental factors likely play a role in manifesta- described as the only mechanism through which the anti-
tion of disease and severity. Several genes have been genic material would have a chance to be presented, but
implicated in both SLE and lupus nephritis. Other factors, recent studies have shown that other types of cell death
such as a higher incidence of SLE in monozygotic twins, could also potentially contribute [16]. Usually, the rapid
and greater risk of developing the disease if family mem- clearance of a dead cell would prevent autoantibody for-
bers have SLE or other autoimmune conditions, suggest a mation. However, in patients with SLE, it is possible that
genetic predisposition. either inappropriate clearance of cellular debris or an
Lupus nephritis is a severe complication of SLE that abnormal increase in cell death provides ample opportunity
carries a high morbidity and mortality if left untreated. An for nucleosomes to become antigenic material for autoan-
abnormal urinalysis with or without elevated plasma cre- tibody formation [3, 13, 16]. These autoantibodies can
atinine is present in many patients with LN and SLE, with form immune complexes, which deposit in various organ
proteinuria being a characteristic feature of lupus nephritis systems, leading to dysfunction. There is also a lower
[6]. Several types of renal disease may be found in patients threshold of immune response to the Type I interferon
with SLE, including immune complex mediated glomerular pathway, which has been identified as a potential factor in
disease. These types are differentiated based on SLE [17, 18].
histopathology obtained via renal biopsy [10]. In a few The role of long-lived memory plasma cells and B-cell
cases, patients may have ‘‘silent lupus nephritis,’’ which hyperactivity has been recently elucidated in the patho-
refers to histopathologic disease in the absence of clinical genesis of SLE [19, 20]. The loss of B-cell tolerance to
manifestations. self-antigens and subsequent autoimmune responses lead to
Even with treatment, renal involvement significantly the deposition of circulating immune complexes (CIC)
affects the survival and quality of life of patients with SLE [21]. The size of immune complexes, charge, local hemo-
[11]. Although several clinical trials have evaluated the dynamic factors, and clearing mechanisms of the mesan-
efficacy of treatment options, the ideal regimen remains to gium all influence where the immune complexes localize
be elucidated. Controlled randomized trials on severe within the glomerulus. Renal deposition of autoantibodies
proliferative and membranous lupus nephritis have aimed forms immune complexes, which is the source of renal
to achieve long intervals of remission while attempting to dysfunction in lupus nephritis. This causes the activation of
minimize side effects of therapy. However, some patients the complement system and subsequent release of cytoki-
have disease refractory to these treatments and may benefit nes, leading to renal injury [13, 22]. Studies have also
from new immunomodulatory agents that are currently shown that direct binding of anti-DNA antibodies to
being studied [12]. In this paper, we review the patho- mesangial cells causes an inflammatory response as well as
genesis, classification, clinical manifestations, and the further cellular proliferation [3]. Autoantibodies against
current treatment options for lupus nephritis, including the C1q have also been isolated. These autoantibodies have
newer immunomodulatory agents and therapies. been identified to cause functional C1 deficiency, con-
tributing to defective clearance of cellular debris from
apoptosis and immune complexes [13]. This may further
Pathogenesis fuel the formation of more autoantibodies as nuclear rem-
nants are not adequately removed.
The pathogenesis of systemic lupus erythematosus is roo- Genetic factors play a major role in the pathogenesis of
ted in the formation of autoantibodies that target self DNA lupus nephritis. Predisposing loci include HLA-DR2,
or other self-nuclear antigens. Although not completely HLA-DR3, HLA-DRB1 loci, HLA-DRB*0301, and HLA-
understood, it is suggested that these autoantibodies are a DRB1*1501. However, multiple genes are involved,
result of abnormalities in the clearance of cellular debris including those that regulate immunity and lymphocyte
that result from cellular apoptosis [13]. The formation of signaling. In short, a single gene polymorphism that leads
autoantibodies will eventually cause a loss of immune to SLE has not been identified; rather a combination of
tolerance and further progression of disease [14]. Once a susceptibility genes and/or absence of protective genes are
source of immunogenic material, such as chromatin, is necessary to lead to overt disease [23]. Environmental
utilized to form autoantibodies, a cascade of other factors have also been implicated in the pathogenesis of
autoantibodies are created, and eventually autoantibodies SLE. These include viruses such as EBV, ultraviolet light,
against dsDNA form. silica, dust, and allergies to medications [24].

123
Clin Exp Nephrol (2016) 20:1–13 3

Classification patients may present with a membranous or proliferative


glomerulosclerosis [13]. In one study, kidney biopsy was
The deposition of immune complexes and subsequent offered to SLE patients over an 11-year period; 86 out of
inflammation leads to the glomerulonephritis seen in lupus 195 patients had no clinical renal disease. Of these 86
nephritis patients. Glomerular injury has been classified patients, 15 % were found to have Class III or IV LN, and
into three distinct patterns—mesangial, endothelial, and 10 % had Class V disease on biopsy [28].
epithelial [25]. As with other glomerular diseases, lupus
nephritis may encompass multiple areas of involvement Tubulointerstitial disease
and the histopathology may be pleomorphic, which com-
plicates both clinical syndromes and the classifications of Some patients with SLE may have tubulointerstitial dis-
severity for glomerular injury [13, 25]. Kidney biopsy ease, manifested by immune deposits of immunoglobulins
remains the gold standard for diagnosis of lupus nephritis. or complements along the basement membranes of tubules
The World Health Organization (WHO), the International and interstitial capillaries [29]. This is more commonly
Society of Nephrology, and the Renal Pathology Society found in patients with Class III or IV LN, often concur-
(ISN/RPS) classify lupus nephritis based on glomerular rently with glomerular disease.
pathology [13]. Advances in technology and research have
allowed both classification systems to be revised on many End-stage renal disease due to lupus nephritis
occasions and the most recent version of classification will
be reviewed. Classes I through VI are described in Table 1. End-stage renal disease (ESRD) due to LN is characterized
by fibrosis on histology [13]. At this stage, dialysis is
Silent lupus nephritis necessary and ultimately, kidney transplantation is required
for continued survival [27]. The percentage of patients who
Lupus nephritis can be classified as ‘‘silent’’ if the patient is progress to end-stage LN varies from 5 to 50 % [29]. These
asymptomatic yet renal biopsy demonstrates evidence of patients have increased mortality during the first several
nephritis [26]. Despite no clinical signs of disease, a few months of dialysis due to increased rate of infections and

Table 1 Histopathological and glomerular classification of lupus nephritis


Class Primary location of Microscopy
immunocomplexes

I: Minimal Mesangium Minimal deposition of mesangium and often appears normal on light microscopy. Diagnosed
Mesangial based on immunofluorescence or electron microscopy (EM)
Weening et al.
[25]
II: Mesangial Mesangium Defined by mesangial hypercellularity. Deposition visualized through light microscopy. May
Proliferative have minimal deposition of subendothelium or subepithelium on EM
Weening et al.
[25]
III: Focal Subendothelium Endocapillary or extracapillary glomerulonephritis comprising \ 50 % of glomeruli.
Weening et al. Characterized by focal subendothelial deposits, with variable involvement of the mesangium.
[25] Further subdivision based on whether lesions are active (A) or chronic (C)
IV: Diffuse Subendothelium Endocapillary or extracapillary glomerulonephritis comprising [50 % of glomeruli.
Weening et al. Characterized by diffuse subendothelial deposits, with variable involvement of the
[25] mesangium. Subdivision of class based on percentage of lesions that are segmental (involving
less than 50 % of glomerular tuft) or global (involving more than 50 % of glomerular tuft).
Further subdivision based on active (A) or chronic (C) lesions, as seen in class III
V: Membranous Subepithelium Subepithelial immune deposits seen on light microscopy. Can be present with Class III or IV
Weening et al. disease
[25]
VI: Advanced Variable [90 % of glomeruli globally sclerosed due to advancement of Class III, IV, or V disease. No
Sclerosing active inflammation
Weening et al.
[25]

123
4 Clin Exp Nephrol (2016) 20:1–13

underlying immunosuppression from immunosuppressant there is no expert consensus on what constitutes remission,
medications. Long-term outcomes for SLE patients with flare, and response [43]. In some studies, remission has
ESRD due to LN are similar to other patients with chronic been defined as less than 10 red blood cells/high-power
kidney disease (CKD) stage V [29]. field and less than 1 g protein in a 24-h urine sample, in the
absence of doubling of serum creatinine level [11]. This is
Clinical manifestations further discussed in the treatment section under therapies
for specific classes of lupus nephritis.
Renal involvement in SLE patients often develops con- Renal biopsy findings are essential in guiding treatment.
currently or soon after diagnosis. Patients may experience Immunosuppressive therapy is recommended for patients
periods of remissions and exacerbations. Clinical involve- with focal or diffuse proliferative lupus nephritis, and in
ment often correlates with glomerular involvement of dis- certain patients with membranous LN, especially those
ease. Class I patients have almost no, or minimal evidence with nephrotic syndrome, an elevated serum creatinine and
of kidney disease, and Class II patients may have minimal concurrent proliferative disease [43]. Progression of lupus
clinical involvement. Class III disease, focal proliferative nephritis, like chronic kidney disease in general, is partly
LN, is often associated with active lupus disease and related to intra-glomerular hypertension. Hypertension
serologies. These patients tend to have hypertension, pro- should be aggressively managed and patients with pro-
teinuria more than 1 g/day, elevated creatinine, and/or teinuria should receive renin-angiotensin system blockade
nephrotic syndrome. Patients with Class IV disease, diffuse therapy (angiotensin-converting enzyme inhibitor or
proliferative LN, tend to present with the most active and angiotensin receptor II blocker), and lipid-lowering therapy
severe clinical manifestations. In addition to findings [43]. In particular, all patients with proteinuria of more
associated with Class III disease, they typically present than or equal to 0.5 g in 24 h should receive renin-an-
with high anti-DNA antibody titers, low serum comple- giotensin system blockade [13]. With regards to lipid
ment levels, active urinary sediment with erythrocytes and lowering, therapy, the SHARP (Study of Heart and Renal
cellular casts, and nephrotic syndrome [29]. Patients with Protection) trial did not reveal a risk reduction of pro-
Class V disease, membranous LN, usually present with gression to ESRD with simvastatin/ezetimibe therapy [45].
nephrotic syndrome and have an increased risk of throm- However, lipid-lowering therapy should be considered in
bosis. Patients with Class VI disease, end-stage LN, have patients with lupus nephritis because of its significant
hypertension, a decreased glomerular filtration rate, and cardiovascular benefit.
have often had years of lupus flares leading to renal
sclerosis. Treatment of class I and II lupus nephritis

Patients with class I and II LN generally have a very good


Treatment prognosis. The treatment strategy consists of regular
monitoring for disease activity and optimal blood pressure
Although there have been several randomized controlled control. Treatment for Class II LN (mesangial-proliferative
trials comparing various therapies (Table 2), the optimal LN) patients with proteinuria \1 g/days is dictated by the
treatment of patients with LN remains to be elucidated [21, extra-renal clinical manifestations of lupus [43, 44]. Those
30–39]. with proteinuria [3 g/days are treated with corticosteroids
Lupus nephritis progresses to end-stage kidney disease or calcineurin inhibitors, similar to patients with minimal
in 5–10 % of patients 10 years after diagnosis [38]. change disease. The lowest risk for CKD progression was
Without treatment, the overall prognosis is very poor, found in patients whose systolic blood pressure was
especially for proliferative LN. Patients with WHO Class maintained in a range of 110–129 mmHg, especially in
IV lupus nephritis have a reported 5-year survival of 17 % those individuals with proteinuria [1 g/days [45]. An
without intensive immunosuppressive treatment [6]. Cur- increase in relative risk for CKD progression at pressures
rent immunosuppressive therapies have substantially above 130 mmHg has been noted [45]. These patients do
improved outcomes; cyclophosphamide-based regimens, well with angiotensin-converting enzyme (ACE) inhibitors
usually given with concomitant corticosteroids, have or angiotensin receptor blockers (ARB), as these agents not
improved survival to 90 % in many cases [6, 40, 41]. Even only help maintain a blood pressure less than
though remission is induced in a significant proportion of 130/80 mmHg, but also reduce proteinuria and may even
patients, risk of relapse remains considerably high and has prevent the development of biopsy proven glomeru-
been reported to range from 18 to 46 % [42]. lonephritis [46, 47]. As per the eighth Joint National
According to the 2012 American College of Rheuma- Commission guidelines, which synthesized data from three
tology guidelines for the management of lupus nephritis, main trials (MDRD, AASK, and REIN 2), the optimal

123
Table 2 Randomized controlled trials on therapies for lupus nephritis
Authors Trial Age sample Diagnostic criteria Exclusion criteria Comparison Outcome
(year) type criteria size

Lewis RCTa C16 86 Lupus diagnosed by ARAb Pregnancy Prednisone and cyclophosphamide vs Addition of plasmapheresis did not
et al. criteria Creatinine Prednisone and improve prognosis of patients with
[30] WHO Class III, IV, V based concentration [6 mg/dl cyclophosphamide ? plasmapheresis SLE
on renal biopsy Previous treatment with
plasmapheresis
History of myocardial
Clin Exp Nephrol (2016) 20:1–13

disease
History of malignancy
Peptic ulcer disease and
active liver disease
Chan RCT N/A 42 Lupus diagnosed by ARA Serum creatinine MMFc ? prednisolone vs MMF ? prednisolone has same
et al. criteria concentration [3.4 mg/ Prednisolone ? cyclophosphamide then efficacy as
[31] WHO Class IV based on renal dl changed to AZAd Prednisolone ? cyclophosphamide
biopsy Severe infection or then changed to AZA
cerebral lupus
Poor compliance
Pregnancy
Treatment with
cyclophosphamide
within last 6 months
Treatment with
prednisolone at 0.8 mg/
kg/day for [2 weeks
Houssiau RCT C14 90 Lupus diagnosed by ACRe Take cyclophosphamide High-dose IV cyclophosphamide ? AZA vs No statistical significance between
et al. criteria or AZA during previous Low dose IV cyclophosphamide ? AZA the two modalities; good clinical
[32] WHO Class III, IV, Vc, Vd year results may be possible with just
Renal thrombotic low dose treatment
microangiopathy
Preexisting chronic renal
failure
Pregnancy
Previous malignancy
Diabetes mellitus
Poor compliance
5

123
6

Table 2 continued
Authors Trial Age sample Diagnostic criteria Exclusion criteria Comparison Outcome
(year) type criteria size

123
Yee et al. RCT 16–65 32 Lupus diagnosed by ACR Treatment with Intermittent pulse cyclophosphamide and No significant difference in efficacy
[33] criteria cyclophosphamide or methylprednisolone vs. Oral continuous between two treatment modalities
WHO Class III, IV based on AZA 3 weeks prior cyclophosphamide and prednisolone for tested
renal biopsy Pure mesangial or pure 3 months followed by continuous AZA and
membranous- prednisolone
proliferative
glomerulonephritis on
biopsy
Previous treatment with
cyclophosphamide
for [3 months
Previous malignancy
Non-lupus renal disease
Contreras RCT C18 59 Lupus diagnosed by ARA Creatinine IV cyclophosphamide followed by oral MMF vs. Maintenance with oral AZA or oral
et al. criteria clearance \20 ml/min IV cyclophosphamide followed by oral AZA MMF were more efficacious than
[34] WHO Class III, IV, or Vb Significant infection vs. long-term IV cyclophosphamide long-term IV cyclophosphamide
based on renal biopsy therapy
Pregnancy
[7 doses of IV
cyclophosphamide
Treatment with
AZA [8 weeks
Chan RCT N/A 62 Lupus diagnosed by ARA Serum creatinine MMF vs CTX-AZA as continuous induction- MMF has same efficacy as CTX-
et al. criteria concentration [4.52 mg/ maintenance treatment AZA as continuous induction-
[35] WHO Class IV based on renal dl maintenance treatment
biopsy Severe infection or
cerebral lupus
Treatment with CTX or
MMF within 6 months
prior to study
Treatment with
prednisolone at
doses [0.4 mg/kg per
day for [2 weeks
Clin Exp Nephrol (2016) 20:1–13
Table 2 continued
Authors Trial Age sample Diagnostic criteria Exclusion criteria Comparison Outcome
(year) type criteria size

Moroni RCT C16 75 Lupus diagnosed by ACR Silent nephritis Cyclosporine vs. AZA in maintenance therapy No significant difference in efficacy
et al. criteria Renal diseases unrelated between two treatment modalities
[36] WHO Class IV, Vc, or Vd to lupus tested
based on renal biopsy Treatment with
cyclosporine or AZA
6 months prior to
Clin Exp Nephrol (2016) 20:1–13

screening visit
Cumulative
cyclophosphamide dose
of [200 mg/kg
-Previous malignancy
Appel RCT 12–75 370 Lupus diagnosed by ACR Insufficient proteinuria MMF vs IV cyclophosphamide as induction MMF was not statistically superior to
et al. criteria Serious infection or illness treatment IV cyclophosphamide as induction
[37] WHO Class III, IV, V based Prohibited concurrent
on renal biopsy medication
Pregnancy
Leukopenia
Renal biopsy not met time
criteria
Houssiau RSTf C14 105 Lupus diagnosed by ACR Treatment with AZA vs MMF as long-term immunosuppressive MMF was not statistically superior to
et al. criteria glucocorticoids in the treatment of LN AZA
[38] WHO Class III, IV, Vc, Vd month prior
based on renal biopsy Treatment with
cyclophosphamide,
AZA, MMF, or
cyclosporin A in
previous year
Non-lupus renal disease
Dooley RCT 12–75 227 Active class III, IV, or V LN N/A Oral MMF vs Oral AZA ? placebo MMF superior to AZA in maintaining
et al. with prior response to oral renal response and in preventing
[39] MMF or IV relapse
cyclophophamide during
induction study
7

123
8 Clin Exp Nephrol (2016) 20:1–13

blood pressure goal for all ages with CKD is \140/90 and

Adding rituximab is not superior to


use of ACE inhibitors and ARBs is essential to achieve this
goal, unless a contraindication is present [48–50]. How-

just MMF ? corticosteroids


ever, recent studies have warned against the use of ACE
inhibitors and ARBs in combination due to a higher risk of
complications.

Hydroxychloroquine
Outcome

Antimalarials, such as hydroxychloroquine, have long been


used as a treatment modality for lupus and recent evidence
suggests that they may have a role in the treatment of lupus
Adding rituximab to MMF ? corticosteroids vs
Adding placebo to MMF ? corticosteroids

nephritis. The Hopkins lupus cohort [51], a prospective


database, found that hydroxycholoroquine increases the
chance for complete renal response in conjunction with
mycophenolate mofetil (MMF) therapy for membranous
lupus nephritis. Of patients on hydroxycholoroquine, 64 %
achieved remission within a year compared with only
22 % of those not on hydroxychloroquine (p = 0.036, log-
rank test) [52]. In a Canadian study, hydroxycholoroquine
reduced lupus nephritis flare incidence by 74 %, but did
not reach statistical significance (p = 0.25) [49, 53].
Comparison

Fessler et al. have reported that hydroxychloroquine may


delay or prevent renal damage [52]. All patients with LN
of any class are treated with hydroxychloroquine (maxi-
mum daily dose of 6–6.5 mg/kg ideal body weight) unless
eGFR \ 25 ml/minute/

there is a specific contraindication to the drug. There is


some evidence that hydroxychloroquine may protect
[50 % glomerular

Interstitial fibrosis
Exclusion criteria

against the onset of LN, relapses of LN, and ESRD [54].


Another interesting observation from the Hopkins lupus
sclerosis

1.73 m2

cohort included reduction in thrombosis with hydroxy-


chloroquine usage, particularly for antiphospholipid anti-
body-positive patients [51]. Hydroxychloroquine also has a
favorable impact on lipid profiles [54]. However, it is
supported by renal biopsy

known to be associated with retinal toxicity; thus, patients


Lupus diagnosed by ACR

ISN/RPS Class III or IV

taking Hydroxychloroquine should receive annual eye


exams.
Diagnostic criteria

ANA positivity

Treatment of Class III and IV or proliferative lupus


criteria

nephritis

Proliferative glomerulonephritis (Class III and IV) rep-


American College of Rheumatology

resents the most severe form of lupus nephritis and


sample

American rheumatism association

requires aggressive treatment to prevent progressive loss


size

144

of renal function and to minimize associated morbidity


Randomized-superiority trial
Randomized-controlled trial
criteria

and mortality. Treatment consists of an induction and


16–75

Mycophenolate mofetil
Age

maintenance phase. Partial response to therapy is defined


as a 25 % increase in estimated glomerular filtration rate
Table 2 continued
Trial

RCT
type

(eGFR) from baseline abnormal GFR prior to initiation


Azathioprine

of treatment, a 50 % reduction in urinary proteinuria or


urinary protein creatinine ratio of 0.2–2.0, and a change
Authors

et al.
Rovin
(year)

[21]

from active to inactive urinary sediment. Complete


response is defined as eGFR [90 ml/min/1.73 m2,
b

d
a

123
Clin Exp Nephrol (2016) 20:1–13 9

a [ 50 % reduction in urinary protein-to-creatinine ratio Mycophenolate mofetil (MMF) has been found to be at
of \0.2 and inactive urinary sediment [55]. The key least equivalent to cyclophosphamide for induction ther-
components for both phases include corticosteroids and apy. The Aspreva Lupus Management Study (ALMS) trial
immunosuppressive agents. The average duration of compared MMF to cyclophosphamide in 370 patients with
induction phase in most clinical trials has been lupus nephritis. At 24 weeks, no difference was found
3–6 months with varying maintenance phase durations between the groups with the endpoint of reduction in urine
ranging 2–3 years (MAINTAIN and ALMS trial) [38, protein-to-creatinine ratio to less than three, improvement
39]. The median time to remission is approximately in creatinine or reduction of proteinuria [37]. MMF was
10 months [56]. associated with a higher response rate in Black and His-
panic patients, and a lower incidence of ovarian failure and
Induction therapy alopecia [37].
Although the guidelines list that either cyclophos-
Induction therapy consists of potent immunosuppressive phamide or MMF may be used, some evidence suggests
treatment regimens to achieve remission. This is indicated that one particular agent might be a better option depend-
for patients with diffuse or moderate to severe focal pro- ing on race, disease activity, and patient preference. Black
liferative lupus nephritis. The Kidney Disease: Improving and Hispanic patients may have higher success in achiev-
Global Outcomes and the joint European League Against ing remission with MMF and may require higher induction
Rheumatism and European Renal Association-European doses than patients of other racial/ethnic groups [37, 60].
Dialysis and Transplant Association (KDIGO, EULAR/ Asian patients demonstrated significant intolerance when
ERA-EDTA) guidelines suggest that induction therapy given high daily doses of MMF and thus had higher
should include glucocorticoids and either cyclophos- withdrawal rates in the MMF arm. However, when given
phamide or mycophenolate mofetil (MMF) [43, 44, 57]. lower doses, these patients had better response rates [61].
Within the first 3 months, patients are closely monitored. If In patients with rapidly rising creatinine and/or crescents
there are signs of disease progression such as worsening on renal biopsy indicative of severe LN, cyclophosphamide
serum creatinine or proteinuria, the immunosuppressive is preferred, as it is one of the few drugs that has been
agent may be changed (cyclophosphamide to MMF or vice studied in a randomized control trial and proven to be
versa). effective [62]. Furthermore, it is important for both patient
Glucocorticoid therapy when used alone is not as preference and drug toxicities to be taken into account. For
effective as combined therapy with an immunomodulator. example, a woman desiring children would want to avoid
A meta-analysis found that cyclophosphamide plus gluco- cyclophosphamide given its side effect of ovarian toxicity.
corticoids compared to glucocorticoids alone reduced the Patients on cyclophosphamide should also be monitored for
risk of renal progression as measured by creatinine dou- malignancies, particularly bladder cancer. A yearly Pap
bling, but also increased the risk of ovarian failure in some smear test for cervical cancer screening is recommended,
trials [58]. along with periodic urine studies, serum creatinine, and
There are no set guidelines on oral dosing of steroids but complete blood counts [63]. For patients on glucocorti-
most studies used the following regimen for induction: in coids, checking serum and urine glucose levels as well as
severe disease, intravenous methylprednisolone cholesterol levels is recommended [63].
500–1000 mg given over 30 min daily for 3 days was used, Calcineurin inhibitors such as tacrolimus may be used
then transitioned to oral prednisone tapered from an initial when patients cannot tolerate standard therapies. In a meta-
dose of 0.5–1 mg/kg to a maintenance level of analysis performed by Deng et al. in Chinese populations,
5–10 mg/day, as per NIH protocol [59]. several studies revealed that tacrolimus appears to be a
Cyclophosphamide may be given as a high-dose regi- safer, more efficacious alternative to cyclophosphamide
men (NIH protocol) or lower-dose (Euro-lupus protocol). when used in the induction phase of treatment. Tacrolimus
The higher-dose regimen includes intravenous cyclophos- was found to be superior in causing complete remission,
phamide 0.5–1 g/m2 monthly for 6–7 months followed by while having a more tolerable side effect profile [64].
quarterly administration of the same dose. Alternatively, a Rituximab, an anti-CD20 B-cell agent, has been used in the
lower-dose regimen, which consists of intravenous past for induction therapy. The LUNAR (Lupus Nephritis
cyclophosphamide 0.5 g/m2 (six biweekly pulses) for Assessment with Rituximab) trial, a multinational study in
10 weeks followed by azathioprine, has been can be used. 2012, primarily assessed the efficacy of Rituximab when
The Euro-Lupus trial found that Caucasian patients with added to MMF and steroids. However, there was no sta-
mild to moderate renal disease (mean serum creatinine of tistically significant response with regards to complete
1.15) had similar 10-year outcomes with the lower-dose remission [21]. Further studies may be needed to elucidate
regimen [32]. Rituximab’s role in induction therapy.

123
10 Clin Exp Nephrol (2016) 20:1–13

Multi-targeted Drug Induction Therapy mycophenolate mofetil and prednisolone combination was
found to be just as effective as a regimen of cyclophos-
The multi-targeted drug therapy (MDI) approach, as phamide and prednisolone, each followed by azathioprine
demonstrated by a recent trial performed in China, and prednisolone [31]. Azathioprine is the preferred choice
attempted to target different aspects of inflammation for women in complete remission who desire to become
implicated in LN. The population studied included patients pregnant in the future. Low dose prednisone is continued in
with biopsy proven LN. Tacrolimus, an inhibitor of T-cell patients who are on maintenance therapy, with the goal of
activation, in addition to MMF, an inhibitor of B-cell achieving symptomatic control with the lowest possible
activation, and steroids (these constituted the MDI arm) dose. Although cyclosporine was found to result in a robust
were directly compared to cyclophosphamide and steroids. reduction in proteinuria, creatinine clearance post-treat-
The study found that at low doses, the MDI group achieved ment was lower when compared to that of patients treated
complete remission within 24 weeks as compared to the with cyclophosphamide [67].
cyclophosphamide group. However, there was an increased
rate of serious adverse effects in the MDI arm. These Treatment of Class V or membranous lupus
adverse effects included pneumonia, varicella zoster nephritis
infection, upper respiratory tract infection, skin and soft
tissue infections, epilepsy, septicemia and doubling of Patients with sub-nephrotic proteinuria and pure membra-
creatinine. Moreover, the efficacy and side effect profile in nous lupus nephritis have a good prognosis regardless of
people of other ethnicities remains to be determined, as the treatment options. Pure membranous LN is rare and thus
study was performed only in Chinese patients [65]. randomized control trials to evaluate superiority of treat-
ment modalities are limited. No consensus of management
Maintenance therapy has yet emerged for this group of patients who may not
require any specific therapy beyond renin-angiotensin
Approximately 50 % of LN patients may experience system blockade [68]. Patients with membranous LN with
relapse after induction therapy, which makes maintenance nephrotic range proteinuria do well with use of steroids and
therapy essential for patients with severe LN [44]. addition of immunosuppression. In one randomized con-
Cyclophosphamide was previously the treatment of choice, trolled trial, the remission rates were 27 % with prednisone
based on results of the initial NIH trial. It is however alone, 60 % with addition of cyclophosphamide and 83 %
associated with significant adverse events including infer- with addition of cyclosporine [69]. The response of the
tility, alopecia, and bladder malignancy [66]. cyclosporine group in contrast to the cyclophosphamide
In 2004, Contreras et al. evaluated patients treated with group was faster in terms of reduction in proteinuria but the
induction therapy using cyclophosphamide, followed by latter group had fewer relapses. Subsequent studies have
maintenance therapy with intravenous cyclophosphamide, also demonstrated the efficacy of oral MMF as compared to
oral azathioprine or oral mycophenolate. The study enrol- intravenous cyclophosphamide induction therapy. The
led fifty-nine patients with lupus nephritis (WHO Class III, remission rates, relapses, and overall clinical courses were
IV) who received induction therapy consisting of a maxi- similar with these two agents [70].
mum of 7 monthly boluses of intravenous cyclophos-
phamide plus corticosteroids and subsequently were Biologic therapies
randomized to one of three maintenance therapies: quar-
terly intravenous injections of cyclophosphamide, oral Biologic therapies have recently gained more momentum
azathioprine (1–3 mg/kg of body weight/day), or oral in the treatment of lupus nephritis. The use of rituximab as
mycophenolate mofetil (500–3000 mg/day) for one to an off- label treatment for LN is increasing and while
3 years [34]. The results showed that the 72-month event- clinical experience is lacking in the use of Belimumab,
free survival rate for the composite end point of death or clinical trials have shown favorable response [60, 71].
chronic renal failure was higher in the MMF and azathio- Biologics may lead to significant changes in the manage-
prine groups than in the cyclophosphamide group ment of SLE in the future.
(p = 0.05 and p = 0.009, respectively). Since then, these
two drugs have often been used in maintenance protocols. Belimumab
A meta-analysis, comparing azathioprine to MMF for
maintenance therapy showed no significant difference in Belimumab is a monoclonal antibody against B-Cell acti-
mortality or progression to ESRD. MMF was however vating factor/B-lymphocyte stimulator (Blys), a cytokine
found to have a lower rate of renal relapse [67]. For that releases survival signals for B cells and is often
patients with diffuse proliferative lupus nephritis, a overproduced in SLE patients. Inhibition of this cytokine

123
Clin Exp Nephrol (2016) 20:1–13 11

results in reduced CD20?B lymphocytes and a shorter life favorable outcomes have been noted in patients with dif-
of plasma cells. Belimumab was found to be effective in fuse proliferative disease. Improvement in survival
reducing flares, disease severity and proteinuria in SLE regardless of diagnosis and class has been achieved with
patients [13, 71]. modern immunosuppressive therapies and better supportive
care [29].
Rituximab Lupus nephritis tends to occur more frequently in
African Americans and overall less favorable prognosis is
Rituximab is a chimeric monoclonal antibody to CD20. As also notable within this population. This may relate to
mentioned above, Rituximab was not found to be as genetic and/or socioeconomic factors. Other factors asso-
effective as other traditional therapies for induction or ciated with poor outcomes included age, gender, duration
maintenance. It may however have a role in the treatment of SLE diagnosis, uncontrolled hypertension, anemia, ele-
of patients with refractory disease. A pooled data analysis vated serum creatinine, high rate of decline in GFR, and
of patients with refractory disease showed that Rituximab chronic renal scarring [29]. Retrospective studies have
was particularly effective in patients with mixed mem- shown that 5-year survival rate of these patients depends on
branous-proliferative lupus nephritis and Class III LN. Of whether there is membranous involvement only, or if
note, there have been a few reported cases of progressive proliferative lesions are also present. Those with prolifer-
multifocal leukoencephalopathy, a fatal demyelinating ative lesions in addition to membranous ones had about
disease, with the use of Rituximab [13]. 20–48 % 10-year survival in one study, while those with
only membranous disease had a 72 % survival [29].
Abatacept and other biologics Other factors that may affect clinical course include
adherence to treatment and suboptimal care [72]. Several
Abatacept is a soluble Fc:CTLA-4 fusion protein that interventions have been proposed to improve adherence
competes with CD28 to prevent T-cell activation. A such as repeated explanations regarding why each treat-
phase II/III study with Abatacept plus MMF and corti- ment is prescribed, an overall treatment plan outlined from
costeroids was terminated prematurely due to inefficacy therapy initiation, and the use of pill counters and aids [72].
[13]. Other trials analyzing the use of biologics in SLE Suboptimal care may arise if a multidisciplinary approach
patients have excluded patients with lupus nephritis. to patient care is not achieved.
Newer trials like RING, a European based randomized
controlled trial on Rituximab as a treatment for patients
with refractory LN, are underway [72]. Tabalumab, an Conclusion
anti-Blys monoclonal antibody, is also being studied in
phase II/III trials [71]. Given that the interferon pathway Over the past decade, advances in immunosuppressive
may also be involved in the pathogenesis of SLE, a few therapies have led to substantial improvement in overall
studies with anti-interferon agents have been undertaken. survival rates of patients with lupus nephritis. This review
Unfortunately, none have shown clinical efficacy as of describes established and new therapies for LN. Table 2
yet [71]. highlights the major trials assessing various therapies
including cyclophosphamide, azathioprine, mycofenolate
mofetil, and dosing regimens.
Clinical course and prognosis For all patients with LN, it is crucial to optimize risk
factors such as proteinuria and hypertension with renin-
The clinical course of patients with LN varies, with up to angiotensin-receptor blockade therapy. When deciding on a
60 % of patients developing progressive kidney failure. particular therapy, it is important to consider potential
Prognosis has improved over the years with the advent of adverse effects such as atherosclerosis, infertility, and bone
new therapies. Patients with kidney injury limited to the disease. Treatment should be individualized based on the
renal mesangium have a good prognosis and are less likely patient’s clinical presentation, biopsy findings, tolerability
to develop progressive renal failure. Those with minimal and long-term goals. Future studies, some of which are
and limited glomerular injury generally have adequate underway, may help shed light on new and refined thera-
response to therapy, and less than 5 % of these patients pies for this condition.
progress to renal failure [29]. Class III patients may
Compliance with ethical standards
advance to Class IV disease. Those patients with more
proliferation, necrotizing features on histopathology, or Conflict of interest The authors declare no conflict of interest. No
crescent formation have a worse prognosis. The least funding was received.

123
12 Clin Exp Nephrol (2016) 20:1–13

References 22. Pisetsky DS, Ullal AJ. The blood nucleome in the pathogenesis of
SLE. Autoimmun Rev. 2010;10(1):35–7.
1. Pollak VE, Pirani CL, Schwartz FD. The natural history of the 23. Boackle SA. Advances in lupus genetics. Curr Opin Rheumatol.
renal manifestations of systemic lupus erythematosus. J Lab Clin 2013;25(5):561–8.
24. Cooper GS, Dooley MA, Treadwell EL, St Clair EW, Gilkeson
Med. 1964;63:537–50.
2. Maroz N, Segal MS. Lupus nephritis and end-stage kidney dis- GS. Risk factors for development of systemic lupus erythe-
ease. Am J Med Sci. 2013;346(4):319–23. matosus: allergies, infections, and family history. J Clin Epi-
3. Pateinakis P, Pyrpasopoulou A. Targeting the B-cell pathway in demiol. 2002;55(10):982–9.
25. Weening JJ, D’Agati VD, Schwartz MM, Seshan SV, Alpers CE,
lupus nephritis: current evidence and future perspectives. Sci
World J. 2013;2013:745239. Appel GB, et al. The classification of glomerulonephritis in
4. Hsu CY, Chiu WC, Yang TS, Chen CJ, Chen YC, Lai HM, et al. systemic lupus erythematosus revisited. J Am Soc Nephrol JASN.
Age- and gender-related long-term renal outcome in patients with 2004;15(2):241–50.
lupus nephritis. Lupus. 2011;20(11):1135–41. 26. Gonzalez-Crespo MR, Lopez-Fernandez JI, Usera G, Poveda MJ,
5. Ortega LM, Schultz DR, Lenz O, Pardo V, Contreras GN. Gomez-Reino JJ. Outcome of silent lupus nephritis. Semin
Review: lupus nephritis: pathologic features, epidemiology and a Arthritis Rheum. 1996;26(1):468–76.
guide to therapeutic decisions. Lupus. 2010;19(5):557–74. 27. Stone JH. End-stage renal disease in lupus: disease activity,
6. Cameron JS. Lupus nephritis. J Am Soc Nephrol JASN. dialysis, and the outcome of transplantation. Lupus.
1999;10(2):413–24. 1998;7(9):654–9.
7. Ippolito A, Petri M. An update on mortality in systemic lupus 28. Wakasugi D, Gono T, Kawaguchi Y, Hara M, Koseki Y, Kat-
sumata Y, et al. Frequency of class III and IV nephritis in sys-
erythematosus. Clin Exp Rheumatol. 2008;26(5 Suppl 51):S72–9.
8. Anaya JM, Canas C, Mantilla RD, Pineda-Tamayo R, Tobon GJ, temic lupus erythematosus without clinical renal involvement: an
Herrera-Diaz C, et al. Lupus nephritis in Colombians: contrasts analysis of predictive measures. J Rheumatol. 2012;39(1):79–85.
and comparisons with other populations. Clin Rev Allergy 29. Kelly WN ea. Clinical Features of SLE. Textbook of Rheuma-
Immunol. 2011;40(3):199–207. tology. Philadephia: WB Saunders; 2000.
9. Varela DC, Quintana G, Somers EC, Rojas-Villarraga A, Espi- 30. Lewis EJ, Hunsicker LG, Lan SP, Rohde RD, Lachin JM. A
nosa G, Hincapie ME, et al. Delayed lupus nephritis. Ann Rheum controlled trial of plasmapheresis therapy in severe lupus
Dis. 2008;67(7):1044–6. nephritis. The lupus nephritis collaborative study group. New
10. Grande JP, Balow JE. Renal biopsy in lupus nephritis. Lupus. Engl J Med. 1992;326(21):1373–9.
1998;7(9):611–7. 31. Chan TM, Li FK, Tang CS, Wong RW, Fang GX, Ji YL, et al.
11. Houssiau FA. Therapy of lupus nephritis: lessons learned from Efficacy of mycophenolate mofetil in patients with diffuse pro-
liferative lupus nephritis. Hong Kong-guangzhou nephrology
clinical research and daily care of patients. Arthritis Res Ther.
2012;14(1):202. study group. New Engl J Med. 2000;343(16):1156–62.
12. Mok CC. Therapeutic options for resistant lupus nephritis. Semin 32. Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, Garrido
Arthritis Rheum. 2006;36(2):71–81. Ed, Ede R, Danieli MG, et al. Immunosuppressive therapy in
lupus nephritis: the Euro-Lupus Nephritis Trial, a randomized
13. Borchers AT, Leibushor N, Naguwa SM, Cheema GS, Shoenfeld
Y, Gershwin ME. Lupus nephritis: a critical review. Autoimmun trial of low-dose versus high-dose intravenous cyclophos-
Rev. 2012;12(2):174–94. phamide. Arthritis Rheum. 2002;46(8):2121–31.
14. Schwartz N, Goilav B, Putterman C. The pathogenesis, diagnosis 33. Yee CS, Gordon C, Dostal C, Petera P, Dadoniene J, Griffiths B,
and treatment of lupus nephritis. Curr Opin Rheumatol. et al. EULAR randomised controlled trial of pulse cyclophos-
2014;26(5):502–9. phamide and methylprednisolone versus continuous cyclophos-
15. Berden JH, Licht R, van Bruggen MC, Tax WJ. Role of nucle- phamide and prednisolone followed by azathioprine and
osomes for induction and glomerular binding of autoantibodies in prednisolone in lupus nephritis. Ann Rheum Dis. 2004;63(5):525–9.
lupus nephritis. Curr Opin Nephrol Hypertens. 34. Contreras G, Pardo V, Leclercq B, Lenz O, Tozman E, O’Nan P,
1999;8(3):299–306. et al. Sequential therapies for proliferative lupus nephritis. New
16. Colonna L, Lood C, Elkon KB. Beyond apoptosis in lupus. Curr Engl J Med. 2004;350(10):971–80.
35. Chan TM, Tse KC, Tang CS, Mok MY, Li FK. Long-term study
Opin Rheumatol. 2014;26(5):459–66.
17. Elkon KB, Stone VV. Type I interferon and systemic lupus of mycophenolate mofetil as continuous induction and mainte-
erythematosus. J Interferon Cytokine Res Off J Int Soc Interferon nance treatment for diffuse proliferative lupus nephritis. J Am
Cytokine Res. 2011;31(11):803–12. Soc Nephrol JASN. 2005;16(4):1076–84.
18. Crow MK. Advances in understanding the role of type I inter- 36. Moroni G, Doria A, Mosca M, Alberighi OD, Ferraccioli G,
ferons in systemic lupus erythematosus. Curr Opin Rheumatol. Todesco S, et al. A randomized pilot trial comparing cyclosporine
2014;26(5):467–74. and azathioprine for maintenance therapy in diffuse lupus
19. Cheng Q, Mumtaz IM, Khodadadi L, Radbruch A, Hoyer BF, nephritis over four years. Clin J Am Soc Nephrol CJASN.
Hiepe F. Autoantibodies from long-lived ‘memory’ plasma cells 2006;1(5):925–32.
of NZB/W mice drive immune complex nephritis. Ann Rheum 37. Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D,
Dis. 2013;72(12):2011–7. Jayne D, et al. Mycophenolate mofetil versus cyclophosphamide
for induction treatment of lupus nephritis. J Am Soc Nephrol
20. Ripoll E, Merino A, Herrero-Fresneda I, Aran JM, Goma M,
Bolanos N, et al. CD40 gene silencing reduces the progression of JASN. 2009;20(5):1103–12.
experimental lupus nephritis modulating local milieu and sys- 38. Houssiau FA, D’Cruz D, Sangle S, Remy P, Vasconcelos C,
temic mechanisms. PLoS One. 2013;8(6):e65068. Petrovic R, et al. Azathioprine versus mycophenolate mofetil for
long-term immunosuppression in lupus nephritis: results from the
21. Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, San-
chez-Guerrero J, et al. Efficacy and safety of rituximab in patients MAINTAIN Nephritis Trial. Ann Rheum Dis.
with active proliferative lupus nephritis: the lupus nephritis 2010;69(12):2083–9.
assessment with rituximab study. Arthritis Rheum. 39. Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy
2012;64(4):1215–26. D, et al. Mycophenolate versus azathioprine as maintenance

123
Clin Exp Nephrol (2016) 20:1–13 13

therapy for lupus nephritis. New Engl J Med. erythematosus clinical trials. Arthritis Rheum.
2011;365(20):1886–95. 2006;54(2):421–32.
40. Mok CC, Ho CT, Chan KW, Lau CS, Wong RW. Outcome and 56. Ioannidis JP, Boki KA, Katsorida ME, Drosos AA, Skopouli FN,
prognostic indicators of diffuse proliferative lupus glomeru- Boletis JN, et al. Remission, relapse, and re-remission of prolif-
lonephritis treated with sequential oral cyclophosphamide and erative lupus nephritis treated with cyclophosphamide. Kidney
azathioprine. Arthritis Rheum. 2002;46(4):1003–13. Int. 2000;57(1):258–64.
41. Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, de 57. Bertsias GK, Tektonidou M, Amoura Z, Aringer M, Bajema I,
Ramon Garrido E, Danieli MG, et al. The 10-year follow-up data Berden JH, et al. Joint European League Against Rheumatism
of the Euro-Lupus Nephritis Trial comparing low-dose and high- and European Renal Association-European Dialysis and Trans-
dose intravenous cyclophosphamide. Ann Rheum Dis. plant Association (EULAR/ERA-EDTA) recommendations for
2010;69(1):61–4. the management of adult and paediatric lupus nephritis. Ann
42. Ponticelli C, Moroni G. Flares in lupus nephritis: incidence, Rheum Dis. 2012;71(11):1771–82.
impact on renal survival and management. Lupus. 58. Flanc RS, Roberts MA, Strippoli GF, Chadban SJ, Kerr PG,
1998;7(9):635–8. Atkins RC. Treatment for lupus nephritis. The Cochrane database
43. Hahn BH, McMahon MA, Wilkinson A, Wallace WD, Daikh DI, of systematic reviews. 2004(1):CD002922.
Fitzgerald JD, et al. American College of Rheumatology guide- 59. Boumpas DT, Austin HA 3rd, Vaughn EM, Klippel JH, Steinberg
lines for screening, treatment, and management of lupus nephri- AD, Yarboro CH, et al. Controlled trial of pulse methylpred-
tis. Arthritis Care Res. 2012;64(6):797–808. nisolone versus two regimens of pulse cyclophosphamide in
44. KDIGO. KDIGO Clinical Practice Guidelines for Glomerulop- severe lupus nephritis. Lancet. 1992;340(8822):741–5.
nephritis. Kidney Int Suppl; 2012. 60. Chan TM. Recent progress in the treatment of proliferative lupus
45. Jafar TH, Stark PC, Schmid CH, Landa M, Maschio G, de Jong nephritis. Am J Med. 2012;125(7):642–8.
PE, et al. Progression of chronic kidney disease: the role of blood 61. Isenberg D, Appel GB, Contreras G, Dooley MA, Ginzler EM,
pressure control, proteinuria, and angiotensin-converting enzyme Jayne D, et al. Influence of race/ethnicity on response to lupus
inhibition: a patient-level meta-analysis. Ann Intern Med. nephritis treatment: the ALMS study. Rheumatol (Oxford).
2003;139(4):244–52. 2010;49(1):128–40.
46. Duran-Barragan S, McGwin G Jr, Vila LM, Reveille JD, Alarcon 62. Walsh M, James M, Jayne D, Tonelli M, Manns BJ, Hemmelgarn
GS. Angiotensin-converting enzyme inhibitors delay the occur- BR. Mycophenolate mofetil for induction therapy of lupus
rence of renal involvement and are associated with a decreased nephritis: a systematic review and meta-analysis. Clin J Am Soc
risk of disease activity in patients with systemic lupus erythe- Nephrol CJASN. 2007;2(5):968–75.
matosus–results from LUMINA (LIX): a multiethnic US cohort. 63. Schmajuk G, Yazdany J. Drug monitoring in systemic lupus
Rheumatol (Oxford). 2008;47(7):1093–6. erythematosus: a systematic review. Semin Arthritis Rheum.
47. Uchida K, Nitta K. Recent advances in the treatment of lupus 2011;40(6):559–75.
nephritis. Clini Exp Nephrol. 2012;16(2):202–13. 64. Deng J, Huo D, Wu Q, Yang Z, Liao Y. A meta-analysis of
48. Ruggenenti P, Perna A, Loriga G, Ganeva M, Ene-Iordache B, randomized controlled trials comparing tacrolimus with intra-
Turturro M, et al. Blood-pressure control for renoprotection in venous cyclophosphamide in the induction treatment for lupus
patients with non-diabetic chronic renal disease (REIN-2): mul- nephritis. Tohoku J Exp Med. 2012;227(4):281–8.
ticentre, randomised controlled trial. Lancet. 65. Liu Z, Zhang H, Liu Z, Xing C, Fu P, Ni Z, et al. Multitarget
2005;365(9463):939–46. therapy for induction treatment of lupus nephritis: a randomized
49. Wright JT Jr, Bakris G, Greene T, Agodoa LY, Appel LJ, trial. Ann Intern Med. 2015;162(1):18–26.
Charleston J, et al. Effect of blood pressure lowering and anti- 66. Donadio JV Jr, Holley KE, Ferguson RH, Ilstrup DM. Treatment
hypertensive drug class on progression of hypertensive kidney of diffuse proliferative lupus nephritis with prednisone and
disease: results from the AASK trial. JAMA. combined prednisone and cyclophosphamide. New Engl J Med.
2002;288(19):2421–31. 1978;299(21):1151–5.
50. Klahr S, Levey AS, Beck GJ, Caggiula AW, Hunsicker L, Kusek 67. Henderson LK, Masson P, Craig JC, Roberts MA, Flanc RS,
JW, et al. The effects of dietary protein restriction and blood- Strippoli GF, et al. Induction and maintenance treatment of
pressure control on the progression of chronic renal disease. proliferative lupus nephritis: a meta-analysis of randomized
Modification of diet in renal disease study group. New Engl J controlled trials. Am J Kidney Dis Off J Natl Kidney Found.
Med. 1994;330(13):877–84. 2013;61(1):74–87.
51. Fangtham M, Petri M. 2013 update: hopkins lupus cohort. Curr 68. Bomback AS, Appel GB. Updates on the treatment of lupus
Rheumatol Rep. 2013;15(9):360. nephritis. J Am Soc Nephrol JASN. 2010;21(12):2028–35.
52. Fessler BJ, Alarcon GS, McGwin G Jr, Roseman J, Bastian HM, 69. Austin HA 3rd, Illei GG, Braun MJ, Balow JE. Randomized,
Friedman AW, et al. Systemic lupus erythematosus in three controlled trial of prednisone, cyclophosphamide, and cyclos-
ethnic groups: XVI. Association of hydroxychloroquine use with porine in lupus membranous nephropathy. J Am Soc Nephrol
reduced risk of damage accrual. Arthritis Rheum. JASN. 2009;20(4):901–11.
2005;52(5):1473–80. 70. Radhakrishnan J, Moutzouris DA, Ginzler EM, Solomons N,
53. A randomized study of the effect of withdrawing hydroxy- Siempos II, Appel GB. Mycophenolate mofetil and intravenous
chloroquine sulfate in systemic lupus erythematosus. The Cana- cyclophosphamide are similar as induction therapy for class V
dian Hydroxychloroquine Study Group. New Engl J Med. lupus nephritis. Kidney Int. 2010;77(2):152–60.
1991;324(3):150–4. 71. van Vollenhoven RF, Parodis I, Levitsky A. Biologics in SLE:
54. Ruiz-Irastorza G, Ramos-Casals M, Brito-Zeron P, Khamashta towards new approaches. Best Pract Res Clin Rheumatol.
MA. Clinical efficacy and side effects of antimalarials in systemic 2013;27(3):341–9.
lupus erythematosus: a systematic review. Ann Rheum Dis. 72. Houssiau FA, Lauwerys BR. Current management of lupus
2010;69(1):20–8. nephritis. Best Pract Res Clin Rheumatol. 2013;27(3):319–28.
55. The American College of Rheumatology response criteria for
proliferative and membranous renal disease in systemic lupus

123

You might also like