You are on page 1of 12

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/224957300

Recent progress in the treatment of lupus nephritis

Article  in  Modern Rheumatology · May 2012


DOI: 10.1007/s10165-012-0655-4 · Source: PubMed

CITATIONS READS

6 477

4 authors, including:

Antonis Fanouriakis Michail Tzanakakis


"Attikon" University Hospital, Athens, Greece General Hospital of Naxos
79 PUBLICATIONS   252 CITATIONS    21 PUBLICATIONS   24 CITATIONS   

SEE PROFILE SEE PROFILE

Dimitrios T Boumpas
Biomedical Research Foundation
398 PUBLICATIONS   17,069 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Neuropsychiatric SLE View project

Practice internship View project

All content following this page was uploaded by Antonis Fanouriakis on 27 May 2014.

The user has requested enhancement of the downloaded file.


Mod Rheumatol
DOI 10.1007/s10165-012-0655-4

REVIEW ARTICLE

Recent progress in the treatment of lupus nephritis


Antonis Fanouriakis • Eleni Krasoudaki •
Michail Tzanakakis • Dimitrios T. Boumpas

Received: 31 January 2012 / Accepted: 18 April 2012


Ó Japan College of Rheumatology 2012

Abstract The treatment of lupus nephritis has seen sig- therapy with a focus on recent RCTs as well as new bio-
nificant advances during the past decade mainly due to the logic agents under development. Furthermore, we propose
publication of well-designed randomized clinical trials a therapeutic algorithm in an effort to facilitate clinical
(RCTs). The choice of treatment is guided by the histo- decision-making in this gradually changing landscape.
pathologic classification but is also influenced by demo- Upcoming European and American recommendations
graphic, clinical, and laboratory characteristics that allow should provide further clarification.
for the identification of patients at risk for more aggressive
disease. For the induction arm, low-dose cyclophospha- Keywords Biologics  Controlled trials  Lupus nephritis
mide regimens and mycophenolate mofetil have been
validated as alternatives to the established National Insti-
tutes of Health regimen of high-dose cyclophosphamide; Introduction
for the maintenance phase, azathioprine and mycopheno-
late compete for treatment of first choice. Rituximab is During the course of the disease, a patient with systemic
efficacious in real-life clinical practice but ineffective in lupus erythematosus (SLE) will face up to a 60 % risk of
clinical trials. The role of recently approved belimumab in developing glomerulonephritis (LN), a manifestation with
lupus nephritis eagerly awaits further documentation. a significant impact on the quality of life and survival [1].
Aggressive management of comorbid conditions, such as Of these patients, 10–30 % will progress to end-stage renal
hypertension and dyslipidemia, is of utmost importance. disease (ESRD) within 15 years of diagnosis [2]. Despite
Here, we review the latest advances in lupus nephritis considerable advances in treatment in recent years, recent
studies in adult and pediatric SLE patients have found that
the incidence of LN-associated ESRD has increased and
A. Fanouriakis (&)  D. T. Boumpas outcomes have not improved [3, 4].
Department of Rheumatology, Clinical Immunology and For physicians providing care for lupus patients, the last
Allergy, University Hospital of Heraklion, University of Crete decade has been intriguing. Various immunosuppressive
Medical School, 71003 Heraklion, Greece
agents have been studied in an effort to achieve clinical
e-mail: afanouriakis@edu.med.uoc.gr
efficacy in terms of remission of nephritis while minimiz-
E. Krasoudaki ing the deleterious side effects of treatment. In this review,
Department of Nephrology, Venizeleion Hospital of Heraklion, we highlight the most important recent advances in the
Heraklion, Greece
field with a focus on (1) induction and maintenance therapy
M. Tzanakakis of proliferative LN, (2) treatment options in membranous
Department of Nephrology, University Hospital of Heraklion, LN, and (3) randomized controlled trials (RCTs) of new
University of Crete Medical School, Heraklion, Greece biologic agents.
The treatment of severe LN involves a period of inten-
D. T. Boumpas
Institute of Molecular Biology and Biotechnology (FORTH), sive immunosuppressive therapy to prevent immunological
Heraklion, Greece injury (induction therapy), followed by a period of less

123
Mod Rheumatol

aggressive maintenance therapy to maintain the response ESRD and doubling of SCr rates, did not differ between the
and prevent flares and damage accrual. The pioneering two groups, with a median time to remission of 9 months.
studies of the National Institutes of Health (NIH) in the More importantly, albeit not statistically significant, there
1980s established intravenous (IV) cyclophosphamide was a difference in the rate of severe infections in favor of
(CY) in combination with steroids as the ‘‘gold standard’’ the low-dose group. Outcomes in both groups were also
for the treatment of proliferative LN. These RCTs in comparable after 10 years of follow-up, with a mean sur-
patients with severe, proliferative LN established that six vival rate of 92 % for both treatment arms [10]. To the
pulses of IV CY (0.5–1 g/m2) and methylprednisolone investigators’ credit, only six patients were lost to follow-
(MP) on consecutive months, followed by quarterly follow- up during the 10-year period. The same study elegantly
up pulses for 2 years, represented a superior treatment showed that early response to therapy (expressed as a drop
regimen and prevented relapses better than a shorter regi- in proteinuria at 6 months) is a reliable predictor of good
men limited to six doses alone [5, 6]. Extension of the renal outcome even in the long-term [11]. Consequently,
follow-up to a total of 11 years provided further docu- low-dose IV CY may serve as a reasonable alternative
mentation of the higher efficacy of the IV CY–IV MP therapy for selected, white European patients with mild to
combination in the long-term outcome of LN [7]. The same moderately severe LN; whether this can be extrapolated to
set of studies clearly showed that oral CY provides no patients of different origin or with more severe disease
additional benefit over IV CY, while contributing to the remains to be seen.
toxicity of the treatment [8]. Therefore, oral administration
has been practically abandoned. The pro-mycophenolate mofetil movement: pros
The significant toxicity accompanying long-term high- and cons
dose CY therapy and the inadequate response of almost
one-third of the patients has stimulated an intense effort to Three RCTs in the last decade have evaluated the efficacy
find alternative therapies for severe proliferative LN. of mycophenolate mofetil (MMF), a potent inhibitor of
Indeed, although rates were comparable between treatment inosine monophosphate dehydrogenase, in comparison to
arms in the NIH trials, bacterial infections (often necessi- standard NIH IV CY regimens as an induction treatment in
tating hospitalization) and premature amenorrhea are seri- proliferative LN. A common finding from these studies,
ous potential side effects of prolonged CY therapy. Given confirmed by subsequent meta-analyses, is that MMF has
the fact that the majority of patients are women of child- at least comparable—if not superior—efficacy to CY
bearing age, gonadal toxicity in particular poses a serious together with a better safety profile. These observations
issue [rates of sustained amenorrhea in young (B30 years generated an ‘‘enthusiastic movement’’ in favor of MMF,
old) females treated with IV CY range from 12 to 43 %] [7]. with some proclaiming ‘‘the end of the CY era’’. This
enthusiasm overlooked specific shortcomings of the MMF
trials, such as the lack of long-term follow-up as well as the
Alternative choices for induction therapy failure to achieve remission in a significant percentage of
patients.
Low-dose IV CY and the Euro-Lupus project Chan et al. [12] first compared MMF (2 g/day for
6 months followed by 1 g/day for 6 months) to oral CY
The aforementioned toxicity concerns, together with the (2.5 mg/kg for 6 months) followed by AZA (1.5 mg/kg/day
common belief that the disease may be less severe in for 6 months) in a Chinese population with diffuse prolif-
Caucasians, led investigators mainly from Europe to seek erative LN. At 12 months, the two treatments had achieved
alternative, less toxic IV CY protocols. In the Euro-Lupus a similar efficacy, with more than 90 % of the patients of
project, 90 patients with diffuse or focal proliferative LN both treatment groups showing complete or partial remis-
(or membranous plus proliferative), but generally mild sion; amenorrhea, leukopenia, and death occurred only in
disease [mean serum creatinine (SCr) 1.2 mg/dl, mean the CY group. Comparable relapse rates and SCr levels,
proteinuria 3.0 g/day], were randomized to receive either accompanied by fewer infections in the MMF group, were
(1) standard 6 monthly pulses of CY (0.5–1 g/m2) followed found at the 5-year follow-up (with an additional 22
by infusions every third month or (2) a shorter treatment patients) [13]. In a subsequent 6-month study from the
course consisting of 500 mg of IV CY every 2 weeks for USA involving racially diverse patients with either prolif-
six doses (cumulative CY dose, 3 g), followed by azathi- erative or membranous LN, MMF was found to be superior
oprine (AZA) maintenance therapy (2 mg/kg/day) [9]. to monthly IV CY in terms of complete remission (22.5 vs.
Both groups received three pulses of 750 mg IV MP in the 5.8 %; of note, the study was designed as a non-inferiority
start of induction therapy, followed by oral doses of ste- trial), with fewer severe infections and vomiting but more
roids with gradual tapering. Primary end-points, such as frequent diarrheal symptoms [14].

123
Mod Rheumatol

Following these encouraging results, the Aspreva Lupus study). Of note, non-Caucasian race (almost 25 % of study
Management Study (ALMS), one of the largest [n = 370 patients) was an independent strong predictor for deterio-
patients, 27 % of whom had renal impairment with an ration of renal function irrespective of type of therapy.
estimated glomerular filtration rate (GFR) of \60 ml/min/ Together, these data suggest that AZA should be
1.73 m2] and most racially diverse RCTs performed in LN, reserved preferably for young white—but not Black or
sought to demonstrate the potential superiority of MMF Hispanic—female patients with mild proliferative or
over IV CY [15]. The response rates at 6 months were membranous LN who strongly wish to conceive and avoid
similar for both groups (56 % for MMF, 53 % for IV CY), the gonadal toxicity of CY [22].
and there were no differences in the frequency of adverse
events (total events 96 % for MMF, 95 % for IV CY). Of Rituximab: good in real-life, poor in clinical trials
note, however, the response to MMF was better in certain
race/ethnic groups, such as Blacks and mixed Latin The use of rituximab (RTX) in LN has generated
American (Hispanic) patients. Following the publication of ambivalent feelings in clinicians caring for SLE patients.
the results from these RCTs, three recent meta-analyses Multiple small, uncontrolled observational studies over
have concluded that MMF is as effective as CY [pooled the past decade have demonstrated the clear efficacy of
relative risk (RR) for complete remission ranging from this agent usually in patients with refractory nephritis who
1.60 to 1.61, P value = non-significant (NS)] and tends to have failed conventional immunosuppressive regimens,
have a better safety profile (pooled RR: 0.17 for amenor- including CY and MMF. A meta-analysis of seven
rhea, 0.41–0.65 for leukopenia, 0.77–0.83 for infections) as observational studies found a complete or partial response
induction therapy for proliferative LN [16–18]. rate of 69 %, with the lowest rates of complete response
Where do these data leave us regarding the use of MMF observed in patients with membranous (type V) LN [23].
in proliferative LN? Most certainly, it cannot be believed More recently, pooled data from centers in the UK and
that we are now beyond the CY ‘‘era’’—especially in cases Spain demonstrate complete or partial response rates in
of severe LN, for which only CY has exhibited beneficial about two-thirds (67 %) of 164 patients with LN treated
long-term effects, i.e., beyond 5 years. On the other hand, with RTX, which was used in the majority of cases as the
MMF is a valid option for induction therapy in moderately second-line option in refractory disease or flares [24];
severe cases of proliferative LN, especially in Black and 76 % of these patients also received concomitant CY or
Hispanic patients or when gonadal toxicity is an issue (see MMF. Not surprisingly, the presence of NS or renal
also section on Maintenance therapy). failure at the time of RTX administration was a predictor
of poor prognosis and non-response.
Any room for AZA in the induction phase? Contrary to the promising results of open-label trials,
the LUNAR trial—a randomized, double-blind, phase III,
The 10-year follow-up data of the Dutch Lupus Nephritis multi-center trial comparing RTX and placebo on top of
Study Group have recently been published. This study standard of care (MMF ? tapering doses of steroids) in
originally tested the efficacy of AZA as induction–main- 144 SLE patients with proliferative LN (types III/IV)—
tenance regimen in 87 European patients with proliferative failed to meet its primary end-point [25]. By week 52, no
LN who were randomized to receive pulse IV CY (totally statistically significant differences were observed between
13 pulses in 2 years) ? oral prednisone versus IV MP ? the two groups in terms of complete or partial renal
AZA ? a tapering dose of oral prednisone [19]. After a response [defined by SCr, normalization of urinary sedi-
median follow-up duration of 5.7 years, the rates of dou- ment, and urine protein-to-creatinine (Pr/Cr) ratio]; of
bling of baseline SCr and renal relapses were higher in the note, there were numerically more responders in the RTX
AZA than the IV CY group. Repeat renal biopsy after group (57 vs. 46 %). Along with EXPLORER, the coun-
2 years of treatment in a subset of patients (n = 39) terpart trial in nonrenal lupus, the LUNAR trial has
showed a significant increase in the chronicity index in the received considerable criticism not only for its lack of
AZA arm; however, these histological variables could not power to prove superiority but also for the use of full,
predict long-term renal outcomes, potentially due to the effective doses of established background therapies that
relatively limited number of repeat biopsies [20]. In the may have potentially diluted any effect attributable to
10-year extension of the follow-up, the results were not RTX. Until more data are available, most experts agree
much different [21]. Renal relapses were significantly more that RTX constitutes a valid therapeutic option in patients
common in the AZA ? MP group (38 vs. 10 % in the IV with aggressive disease refractory to conventional treat-
CY group, with a HR of 4.5), as was the sustained doubling ment regimens (as was the case in most open-label trials
of SCr (16 vs. 8 %, although the latter parameter did not as opposed to the milder forms included in the trials) or in
reach statistical significance due to the lack of power of the the prevention of flares.

123
Mod Rheumatol

Other therapeutic agents patients with LN who received MMF (mean dose 2.8 g) and
tacrolimus (mean trough level 4.7 ng/dl) for 2–54 months
Tacrolimus and multi-target therapy [32]. Overall, well-designed randomized studies with a large
number of patients and long-term follow-up are essential in
Tacrolimus, a more potent calcineurin inhibitor than order to allow generalizability of such results and to be able to
cyclosporine A (CsA), has also shown efficacy in LN with draw definite conclusions regarding the use of tacrolimus
a generally favorable toxicity profile. Initial open-label either alone or as part of a combination regimen in the
trials and an RCT exhibited the beneficial effects of ta- induction therapy of LN.
crolimus as induction therapy in both proliferative and
membranous LN [26, 27]. More recently, in a head-to-head Cyclosporine A
comparison with IV CY, Chen et al. [28] randomized 81
Chinese patients with mild active LN (mostly class IV, Uncontrolled studies including a small number of patients
mean proteinuria approx. 400 mg/24 h, mean SCr\1 mg/dl) have demonstrated the efficacy of CsA, the prototypical
to receive either tacrolimus (started at 0.05 mg/kg/day and calcineurin inhibitor, when used in combination with cor-
titrated to trough levels of 5–10 ng/ml) or six pulses of IV ticosteroids in refractory-to-conventional treatment prolif-
CY (0.75 g/m2 in the first pulse, adjusted to 0.5–1 g/m2 erative LN. Initially, a group of Czech investigators
thereafter). Designed as a noninferiority trial, the study prospectively studied 31 LN patients, 13 of whom were
showed higher, although not statistically significant, rates intolerant or refractory to CY, who were treated with CsA
of complete remission and response for the tacrolimus (dose adjusted to trough level 80–120 ng/ml) either as first-
group (52.4 vs. 38.5 % and 90.5 vs. 82.1 %, respectively) or second-line treatment together with prednisone (20 mg/
after 6 months. However, this difference was entirely day and tapered to B10 mg/day) [33]. With a mean follow-
accounted for by the subset of patients with pure mem- up of 85 months, more than 90 % of patients achieved
branous (class V) LN. In terms of safety, tacrolimus was complete remission (proteinuria \1 g/day and improved or
better tolerated than IV CY, with significantly less frequent stabilized renal function); nearly half of these patients
leukopenia and gastrointestinal disturbance. In a similar experienced a renal flare after CsA was withdrawn. Given
6-month open-label trial with 60 Chinese patients com- the considerable concern associated with CsA nephrotoxi-
paring both tacrolimus and MMF to standard IV CY city, long-term therapy with this agent warrants further
therapy, the rates of complete and partial remission were validation.
comparable, with tacrolimus showing a trend towards fas- In a subsequent randomized study from the same group,
ter resolution of proteinuria [29]. While providing proof of 40 patients with newly diagnosed proliferative LN and
concept for the efficacy of tacrolimus as induction therapy mild renal insufficiency were assigned to sequential
for LN, these studies are limited by their short duration of induction and maintenance therapy with either CY or CsA
follow-up (6 months) and relatively small sample size. [34]. The CY regimen included eight pulses of IV CY
Mixed class IV ? V LN (proliferative lesions superim- (10 mg/kg) administered within 9 months, followed by
posed on membranous LN) represents a high-risk histolog- four to five oral CY boluses (10 mg/day in 6- to 8-week
ical subtype with lower remission rates [30]. Tacrolimus has intervals), while CsA was given orally (4–5 mg/kg/day) for
been used in this subgroup of patients as part of a ‘‘multi- 9 months and then tapered to 3.75–1.25 mg/kg/day within
target’’ combination therapeutic regimen. Combination the next 9 months. Oral steroids were started at 0.8 mg/kg
therapy (tacrolimus 4 mg/day ? MMF 1 g/day) was com- with gradual tapering. In the induction part of the regimen,
pared against IV CY (6–9 pulses of 1 g/m2), both used in the same number of patients (5; 24 % for CY and 26 % for
combination with corticosteroids (three daily pulses IV MP CsA, respectively) met the criteria for complete response/
0.5 g/day followed by oral prednisone) [31]. Enrolled remission at 9 months (defined as normal urinary sediment,
patients (n = 40) had normal renal function (estimated proteinuria \0.3 g/24 h and stable serum creatinine level).
GFR 98 ml/min) and a mean urinary protein excretion of Partial response rates also did not differ. At the end of the
4.4 g/24 h. After 6 months, ten patients in the ‘‘multi-tar- maintenance phase (18 months), 14 % patients in the CY
get’’ group versus one patient in the IV CY group achieved group compared to 37 % of patients in the CsA group were
complete remission. The combination therapy was well in remission; another eight (38 %) and 11 (58 %) patients,
tolerated and no major effects were observed. These data respectively, fulfilled the partial response criteria. How-
need to be examined cautiously in view of the limited ever, this difference in response rates in favor of CsA was
follow-up period (9 months), which does not allow for largely attributed to the higher proportion of patients
solid conclusions to be drawn mainly in terms of long-term achieving a urinary protein excretion rate of \0.3 g/24 h
calcineurin inhibitor nephrotoxicity. In this regard, a recent (response criterion of proteinuria; 38 % in CY group vs.
study reported considerable toxicity in five of seven 74 % in CsA group; p = 0.02). In terms of adverse effects,

123
Mod Rheumatol

the frequency of infection-related adverse events were MMF, irrespective of the magnitude of the renal response
similar, but treatment with CsA was associated with a at the end of the induction period. After a minimum of
transient increase in blood pressure and decrease in GFR. 3 years, the two groups did not differ in terms of time-to-
Until more data are available, current evidence suggests renal flare (the primary end-point), number of severe flares,
that CsA may be considered as a therapeutic alternative to renal remission, or doubling of SCr. Hematological cy-
CY in Caucasian patients with mild-to-moderate pro- topenias were more common in the AZA group. On the
teinuric proliferative LN—but with diligent monitoring for contrary, the ALMS study included a total of 227 patients
hypertension and nephrotoxicity. (more than 50 % non-whites) who achieved a clinical
response to either MMF or IV CY in the induction phase, a
pivotal difference from the MAINTAIN study which did
Maintenance therapy not need response as a prerequisite [37]. These patients
were randomized to receive either MMF (target dose
The need for maintenance treatment to decrease the num- 2 g/day) or AZA (target dose 2 mg/kg/day) and were moni-
ber of flares and accrual of renal damage was demonstrated tored for 3 years. A composite index of treatment failure
in the early NIH trials. In these studies, prolonged (defined as either death, ESRD, renal flare, sustained
administration of quarterly pulses of IV CY for 2 years doubling of SCr, or need for rescue therapy) was used as
decreased the rate of relapses compared to a shorter the primary end-point. In the intention-to-treat analysis,
6-month regimen [5]. Although the optimal duration of MMF was superior to AZA with a hazard ratio for treat-
maintenance therapy has yet to be elucidated, most clini- ment failure of 0.44 (p = 0.003) and a failure rate of 16 %
cians will keep their patients on immunosuppression for at as compared to 32 % in the AZA group at 3 years. Of note,
least 2 years after remission has been achieved. For such a Black patients and those who received IV CY during the
long period, CY does not represent a viable option due to induction phase were less likely to experience treatment
its long-term toxicities of alopecia, hemorrhagic cystitis, failure if treated with MMF for maintenance. Similar to the
bladder cancer, gonadal damage, and early menopause. MAINTAIN study, leukopenia was more common in the
Consequently, other agents have been extensively studied AZA group, as were side effects that led to therapy with-
in the last decade in an effort to minimize toxicity without drawal. Table 1 summarizes the similarities and differ-
sacrificing efficacy. ences of the main head-to-head maintenance studies of
The long-awaited recent publication of the maintenance MMF and AZA.
part of the ALMS trial marks the completion of a series of The discrepancy in the results of these two previous
studies comparing the two main agents currently used for studies may be partly explained by differences in the
maintenance therapy in LN—MMF and AZA. Contreras induction protocol, the number and ethnicity of the
et al. [35] initially compared MMF (0.5–3 g/day) and AZA included patients (smaller size, with White patients
(1–3 mg/kg/day) or quarterly pulses of IV CY as mainte- in MAINTAIN, racially diverse large trial in ALMS),
nance therapy in proliferative LN following remission with the prerequisite for response to induction treatment (in
seven IV CY pulses. The study population (59 patients in ALMS), and the outcome measures used (single in
total) mostly comprised patients of Black and Hispanic MAINTAIN, composite in ALMS). Instead of trying to
origin. Over 6 years of follow-up, the rate of survival establish the superiority of one agent over the other, the
without death or chronic renal failure was significantly main conclusion to be drawn is that both AZA and MMF
higher in the MMF and AZA groups than in the CY group. constitute reasonable efficient options for maintenance
Equally important, MMF- or AZA-treated patients devel- therapy of LN. Race, potential for pregnancy (MMF is
oped significantly less amenorrhea, infections, and gastro- associated with an increased risk of spontaneous abortion
intestinal toxicity. The limitations of this study were the and fetal malformations), cost, and availability must be
relatively small number of patients and the use of lower considered in the individual patient to guide selection of
doses of IV CY together with higher doses of corticoste- one drug over the other. As an example, white patients
roids, which may have decreased efficacy and increased the of childbearing age with mild to moderate LN may be
risk for infections. initially treated with AZA, while non-Caucasian older
The MAINTAIN Nephritis Trial included 105 European patients may be candidates for MMF. Contraindications
patients (approx. 80 % Caucasian origin) with moderately or intolerance to one agent may dictate switching to the
severe class III–IV LN who received induction therapy alternative.
with three daily 750 mg IV MP pulses followed by oral All of these data are summarized in Fig. 1, which sug-
prednizolone, and six IV CY pulses of 500 mg at 2-week gests a step-by-step treatment algorithm and choice of
intervals (the Euro-Lupus regimen) [36]. After 3 months, agents for proliferative LN based on baseline risk stratifi-
all patients were randomized to receive either AZA or cation (low-risk vs. high-risk patients)

123
Mod Rheumatol

SCr serum creatinine, Pr protein, WHO World Health Organization, ISN/RPS International Society of Nephrology/Renal Pathology Society, IV intravenous, CY cyclophosphamide, MMF mycophenolate mofetil, AZA azathioprine, HR
Membranous (class V) LN

MMF 15 %–AZA 20 %

MMF 15 %–AZA 32 %

MMF 19 %–AZA 25 %

MMF 16 %–AZA 32 %
HR 0.44 P = 0.003
Class V LN represents a unique subtype of LN. Immune
Treatment failure

HR 0.75 P = ns)
deposits are limited to the subepithelial space and renal
Renal flares

Renal flares

Renal flares
(6 years)

(3 years)

(3 years)
survival rates vary widely, partly owing to caveats in the
Results

original World Health Organization (WHO) classification


of LN, which included biopsies with superimposed prolif-

flare, ESRD, sustained doubling


of SCr, need for rescue therapy,
erative lesions (classes Vc and Vd) along with pure

Time to treatment failure (renal


(sustained doubling of SCr,

membranous changes (Va and Vb) into the same group—


Patient and renal survival

class V LN [38].
Table 1 Major randomized controlled trials comparing mycophenolate mofetil and azathioprine as maintenance therapies in proliferative lupus nephritis

For patients with membranous LN and low-grade pro-


Time to renal flare
Primary end point

ESRD, death)

teinuria (\2 g/day), although there is no consensus, most


experts agree that specific immunosuppressive treatment

death)
may not be necessary and that blockade of the renin–
angiotensin axis (RAAS) is sufficient to prevent progres-
sion to ESRD (risk of approximately 20 % at 10 years). By
monthly for 6 months) or MMF
NIH protocol (0.5–1 g/m2 IV CY

NIH protocol (0.5–1 g/m2 IV CY

Responders only re-randomized

contrast, the presence of significant proteinuria ([2 g/day),


Euro-Lupus (500 mg IV CY
All patients randomized for

All patients randomized for

NS, decreased GFR, or proliferative lesions in the renal


monthly for 6 months)

biweekly for 3 months

for maintenance phase


maintenance phase

maintenance phase

biopsy usually mandate the institution of immunosuppres-


Induction therapy

sive treatment. Unfortunately, the optimal therapeutic regi-


men remains far from certain. Small-sized, open-label
3 g/day

studies have indicated that tacrolimus has beneficial effects


in combination with steroids in membranous LN with
persistent proteinuria [39–41], but these results have to be
Mean proteinuria

validated in larger, more racially diverse randomized trials.


As mentioned above, the ‘‘multitarget’’ therapy with the
combination of prednisone, MMF, and tacrolimus as
(Pr/Cr)

induction therapy for mixed IV ? V LN showed promising


5.2

3.3

4.1

results and could be considered as an alternative regimen


for this subclass of patients, although close monitoring for
Mean SCr

side effects is warranted.


(mg/dl)

In a randomized controlled study, Austin et al. com-


1.7

1.0

1.1

pared CsA (5 mg/kg/day, then adjusted according to


Histologic subtype

changes in SCr), alternate-monthly IV CY (0.5–1 g/m2 9


72 % IV/IV ? V
13 % III/III ? V
classification)

classification)

classification)

6 doses), and glucocorticoids alone in 42 patients with


(ISN/RPS
78 % IV

58 % IV

membranous LN (median GFR 83 ml/min/1.73 m2,


20 % III

31 % III

15 % V
2 % Vb

8 % Vd
3 % Vc
(WHO

(WHO

median proteinuria 5.4 g/day). All patients received


alternate-day oral prednisone (1 mg/kg every other day
79 % white, 12 % Black

44 % white, 10 % Black

for 8 weeks, then tapered to 0.25 mg/kg every other day).


Hispanic/other, 8 %

33 % Asian 13 %
46 % Black, 46 %

Remission rates at 1 year were 27 % with prednisone,


60 % with IV CY, and 83 % with CsA. However, NS
9 % Asian

Hispanic

relapses were significantly less in the IV CY group than in


white

the CsA group (0.2 vs. 2.0/100 patient-months, respec-


Race

hazard ratio, ESRD End-stage renal disease,

tively; P = 0.02). Of note, patients with impaired renal


59 (95 % female)

105 (91 % female)

227 (86 % female)


Number of patients

function at baseline were not randomly assigned to the


CsA arm due to concerns for CsA-induced nephrotoxicity.
This, together with the small number of patients and the
relatively short follow-up limit the generalization of
conclusions. However, apart from the superiority of both
CsA and IV CY over steroid monotherapy for class V LN,
Contreras et al.

Houssiau et al.

Dooley et al.

this study indicates that CsA may require some form of


maintenance therapy (lower CsA doses or other immu-
[35]

[36]

[37]
Study

nosuppressive agent) to prevent relapses.

123
Mod Rheumatol

Fig. 1 Recommended management of proliferative lupus nephritis AZA azathioprine, CY cyclophosphamide, MP methylprednisolone,
based on risk stratification. LN lupus nephritis, SCr serum creatinine, MMF mycophenolate mofetil, IS immunosuppressives, IV intravenous,
Pr/Cr protein-to-creatinine ratio measured in random urine sample (g/g), NR/PR no remission/partial remission

MMF constitutes yet another therapeutic option for the contrary, a recent study demonstrated that the RTX–IV
membranous LN, as it has been shown to exert anti-pro- CY combination markedly improved proteinuria and led to
teinuric effects in this subgroup of patients [42]. Recently, resorption of subepithelial electron-dense deposits in the
a pooled analysis of 84 patients with pure class V LN from basal membrane in six patients with refractory membra-
two aforementioned prospective RCTs comparing MMF to nous LN [45]. Recent data also suggest that RTX might
IV CY as induction therapy in LN (the ALMS and a pre- preserve renal podocytes and provide protection from
vious study by Ginzler et al. [14]) demonstrated that, recurrent focal segmental glomerulosclerosis [46]; the
irrespective of the initial level of proteinuria (nephrotic or ultimate role of RTX in proteinuric membranous LN awaits
subnephrotic), there was no difference between MMF- and further characterization in a larger number of patients.
IV CY-treated patients in terms of percentage change in Collectively, when immunosuppressive therapy is
urine protein and change in SCr [43]. To complicate mat- required in class V LN, the combination of IV CY with IV
ters, MMF monotherapy failed to demonstrate efficacy in a MP, MMF, a course of oral CsA, or tacrolimus, and even
randomized trial for idiopathic membranous nephropathy AZA all constitute acceptable therapeutic options. The
[44]. choice should be individualized and depend on various
A subset of LN patients treated with rituximab in open- factors, including age, race, planning of pregnancy, and
label trials over the last decade had class V LN (20 in cost. In therapy-resistant cases, a course of RTX may be
total). In the previous pooled analysis from European considered. On the contrary, for patients with proteinuria
cohorts, complete and partial response rates were seen in of \2 g/day, a combination of angiotensin converting
two-thirds of patients, albeit complete response was sig- enzyme inhibitors (ACEIs) to minimize urine protein
nificantly less common than that in mixed membranous- excretion with statins and low-dose aspirin may be suffi-
proliferative LN (18 vs. 50 %) [24]. Of note, NS at the time cient. Vigilant monitoring to detect evolution to a more
of rituximab administration predicted a worse response. On aggressive type of nephritis requiring immunosuppressive

123
Mod Rheumatol

treatment (severe membranous or mixed membranous with Adjunct therapy in LN


proliferative LN) is mandatory.
Patients with lupus have an increased risk of developing
atherosclerotic cardiovascular events, attributable not only
Novel therapies to traditional risk factors but also to disease-specific
parameters and the effects of medications [52, 53]. Con-
Biologics and tolerogens: promises and failures sequently, strict control of hypertension, dyslipidemia, and
hyperglycemia is of utmost importance for patients with
The recently approved belimumab (anti-BLyS; B lym- LN.
phocyte stimulator) has not been formally tested in LN. Hypertension remains a clinically important issue as it
However, approximately 15 % of patients participating in has been associated with a worse renal prognosis [54].
two recent phase III studies leading to drug approval (more Most experts recommend that blood pressure must be kept
than 1,500 study participants in total) had renal involve- below 130/80 mmHg for patients with minimal proteinuria
ment at baseline. Pooled data in this subgroup of patients (\0.5 g/day) and below 120/75 mmHg for patients with
showed a positive trend for belimumab in reducing renal significant proteinuria ([1 g/day). Nephritis per se con-
flares (1.4 vs. 2.8 % for placebo) and proteinuria [47]. tributes to hypertension through the RAAS. Consequently,
Although far from conclusive, these results lend support to ACEis or angiotensin receptor blockers are the antihyper-
the future evaluation of BLyS blockade as a therapeutic tensive agents of choice in patients with chronic kidney
option for LN, possibly as add-on therapy in patients with disease owing to their additional antiproteinuric effects;
incomplete response. additionally, in patients with lupus these agents seem to
Abatacept (CTLA4-Ig), a T-cell costimulation modula- delay the development of renal involvement and to be
tor, has been shown to halt disease progression in lupus- associated with an overall lower disease activity [55].
prone mice, especially when combined with CY [48]. Aggressive treatment of dyslipidemia (target low-den-
Following an initial phase IIb trial that failed to show sity lipoprotein \100 mg/dl and triglycerides \150 mg/dl)
efficacy in non-renal SLE (although again on a background is recommended for patients with LN, similar to the high-
therapy of high-dose steroids) [49], two RCTs were laun- risk population for cardiovascular disease. Akin to non-
ched to evaluate abatacept in combination with background SLE patients, statins remain the cornerstone of therapy for
immunosuppression in the treatment of LN. The results of dyslipidemia in the context of LN. Apart from changing the
the first RCT, a 12-month phase II/III trial with MMF ? lipid profile and decreasing overall cardiovascular risk
steroids as the comparator arm, were recently reported in (even in kidney transplant recipients due to LN [56]), there
abstract form [50]. In 298 patients with proliferative LN, is evidence that statins also exhibit anti-inflammatory and
two different doses of abatacept failed to increase the rates immunomodulatory actions [57]. Pravastatin in particular,
of complete response (normalization of GFR, protein when given in combination with RAAS blockade, has also
excretion, and urinary sediment) over placebo, although a been shown to reduce proteinuria, alleviate renal disease,
greater reduction in proteinuria was noted in patients pre- and prolong survival in a mouse model of SLE [58].
senting with NS at baseline. Of note, if less stringent cri- Hydroxychloroquine (HCQ) constitutes a sine qua non
teria for response were applied [as in the ACCESS study for SLE patients. Although usually prescribed to treat
(Abatacept and Cyclophosphamide Combination Therapy musculoskeletal, cutaneous or serosal manifestations of
for Lupus Nephritis), still recruiting participants], complete lupus, antimalarials are gaining ground as an essential
response rates would be higher and significant differences component of LN therapy [59]. Early data from Canada
would be noted among the different arms (34.8 and 29.3 % for showed a 74 % reduction in risk for nephritic flare in
the two dosing regimens of abatacept vs. 21 % in the placebo patients who continued treatment with HCQ as compared
group) [51]. As is common in lupus trials, this discrepancy to those who withdrew from treatment [60]. An observa-
connotes the ‘‘rocky path’’ of choosing clinically meaningful tional study in a multi-ethnic cohort showed a reduced
outcome measures in LN. The full-text report of both RCTs is cumulative probability of renal damage in LN patients
needed in order to draw more solid conclusions for the role of receiving HCQ [61]. Although the study was criticized for
T-cell costimulation modulation in renal SLE. potentially overestimating the protective effects of HCQ, it
Finally, atacicept (TACI-Ig) and ocrelizumab (anti-CD did provide proof-of-concept for the role of the latter in
22)—both targeting B-cells—as well as synthetic tolero- LN. In a small trial (n = 29 patients) of MMF in mem-
gens (such as abetimus sodium or spliceosomal peptide branous LN, concurrent use of HCQ led to a statistically
P140) represent notable recent failures as therapeutic significant improvement in the rate of renal remission at
options in LN either due to unexpected rates of infectious 12 months [62]. The results of cross-sectional studies
complications or to a lack of efficacy [52, 53]. advocate for a beneficial effect of HCQ on serum lipid

123
Mod Rheumatol

levels, but the data are inconsistent and thus the potential outcomes of end-stage renal disease due to lupus nephritis in the
role of antimalarials in dyslipidemia require further vali- US from 1995 to 2006. Arthritis Rheum. 2011;63(6):1681–8.
4. Hiraki LT, Lu B, Alexander SR, Shaykevich T, Alarcon GS,
dation [63]. Solomon DH, et al. End-stage renal disease due to lupus nephritis
Despite optimal therapy, at least 20 % of patients with among children in the US, 1995–2006. Arthritis Rheum. 2011;
LN still progress to ESRD. Recent data show that younger 63(7):1988–97.
patients, African Americans, or Native Americans with LN 5. Boumpas DT, Austin HA 3rd, Vaughn EM, Klippel JH, Steinberg
AD, Yarboro CH, et al. Controlled trial of pulse methylprednis-
receive renal replacement therapy relatively more often; olone versus two regimens of pulse cyclophosphamide in severe
notably, this does not translate into improved survival rates lupus nephritis. Lancet. 1992;340(8822):741–5.
during the first years of treatment [3]. Clinically important 6. Gourley MF, Austin HA, 3rd, Scott D, Yarboro CH, Vaughan
risk factors for the development of ESRD include abnormal EM, Muir J, et al. Methylprednisolone and cyclophosphamide,
alone or in combination, in patients with lupus nephritis. A ran-
SCr values at presentation, delay in treatment initiation, domized, controlled trial. Ann Intern Med. 1996;125(7):549–57.
failure to achieve remission, and uncontrolled systolic 7. Illei GG, Austin HA, Crane M, Collins L, Gourley MF, Yarboro
hypertension [64, 65]. For renal replacement therapy in CH, et al. Combination therapy with pulse cyclophosphamide
lupus patients, retrospective studies carried out during the plus pulse methylprednisolone improves long-term renal outcome
without adding toxicity in patients with lupus nephritis. Ann
last decade support hemodialysis (HD) over continuous Intern Med. 2001;135(4):248–57.
ambulatory peritoneal dialysis (CAPD) due to the lower 8. Austin HA, 3rd, Klippel JH, Balow JE, le Riche NG, Steinberg
incidence of infectious complications (most commonly AD, Plotz PH, et al. Therapy of lupus nephritis. Controlled trial of
peritonitis) and the beneficial effect on disease activity, prednisone and cytotoxic drugs. N Engl J Med. 1986;314(10):
614–9.
although long-term survival rates are similar [66, 67]. 9. Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, Garri-
Kidney transplantation is a viable alternative option for doEdEde R, Danieli MG, et al. Immunosuppressive therapy in
SLE patients with ESRD. In a recent retrospective study lupus nephritis: the Euro-Lupus Nephritis Trial, a randomized
from Korea, patients who underwent kidney transplantation trial of low-dose versus high-dose intravenous cyclophospha-
mide. Arthritis Rheum. 2002;46(8):2121–31.
after 3 years of renal replacement therapy had lower 10. Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, de
complication rates than patients receiving HD and CAPD RamonGarrido E, Danieli MG, et al. The 10-year follow-up data
[68]. Overall patient survival was better in kidney trans- of the Euro-Lupus Nephritis Trial comparing low-dose and high-
plantation than in HD. dose intravenous cyclophosphamide. Ann Rheum Dis. 2010;69(1):
61–4.
11. Houssiau FA, Vasconcelos C, D’Cruz D, Sebastiani GD, de
RamonGarrido E, Danieli MG, et al. Early response to immu-
Conclusion nosuppressive therapy predicts good renal outcome in lupus
nephritis: lessons from long-term followup of patients in the
Euro-Lupus Nephritis Trial. Arthritis Rheum. 2004;50(12):3934–
In recent years, potent drugs, such as MMF, have definitely 40.
enriched our armamentarium in LN, and the future holds 12. Chan TM, Li FK, Tang CS, Wong RW, Fang GX, Ji YL, et al.
great promise with the development of new biologic drugs. Efficacy of mycophenolate mofetil in patients with diffuse pro-
The lupus community is struggling to design optimal ran- liferative lupus nephritis. Hong Kong-Guangzhou Nephrology
Study Group. N Engl J Med. 2000;343(16):1156–62.
domized trials with clinically meaningful end-points in an 13. Chan TM, Tse KC, Tang CS, Mok MY, Li FK. Long-term study
effort to maximize efficacy and minimize toxicity. Ideally, of mycophenolate mofetil as continuous induction and mainte-
advances in our understanding of pathogenesis and phar- nance treatment for diffuse proliferative lupus nephritis. J Am
macogenomics as well as the identification of valid bio- Soc Nephrol. 2005;16(4):1076–84.
14. Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill
markers for monitoring response to treatment will JT, et al. Mycophenolate mofetil or intravenous cyclophospha-
eventually lead to more targeted, individualized therapies. mide for lupus nephritis. N Engl J Med. 2005;353(21):2219–28.
15. Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D,
Conflict of interest None. Jayne D, et al. Mycophenolate mofetil versus cyclophosphamide
for induction treatment of lupus nephritis. J Am Soc Nephrol.
2009;20(5):1103–12.
16. Kamanamool N, McEvoy M, Attia J, Ingsathit A, Ngamjanyaporn P,
References Thakkinstian A. Efficacy and adverse events of mycophenolate
mofetil versus cyclophosphamide for induction therapy of
1. Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla lupus nephritis: systematic review and meta-analysis. Medicine
P, et al. Morbidity and mortality in systemic lupus erythematosus (Baltimore). 2010;89(4):227–35.
during a 10-year period: a comparison of early and late mani- 17. Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Induction and
festations in a cohort of 1,000 patients. Medicine (Baltimore). maintenance therapy for lupus nephritis: a systematic review and
2003;82(5):299–308. meta-analysis. Lupus. 2010;19(6):703–10.
2. Ward MM. Changes in the incidence of end-stage renal disease due to 18. Touma Z, Gladman DD, Urowitz MB, Beyene J, Uleryk EM,
lupus nephritis, 1982–1995. Arch Intern Med. 2000;160(20):3136–40. Shah PS. Mycophenolate mofetil for induction treatment of lupus
3. Costenbader KH, Desai A, Alarcon GS, Hiraki LT, Shaykevich T, nephritis: a systematic review and metaanalysis. J Rheumatol.
Brookhart MA, et al. Trends in the incidence, demographics, and 2011;38(1):69–78.

123
Mod Rheumatol

19. Grootscholten C, Ligtenberg G, Hagen EC, vandenWallBake 36. Houssiau FA, D’Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic
AW, de Glas-Vos JW, Bijl M, et al. Azathioprine/methyl- R, et al. Azathioprine versus mycophenolate mofetil for long-term
prednisolone versus cyclophosphamide in proliferative lupus immunosuppression in lupus nephritis: results from the MAINTAIN
nephritis. A randomized controlled trial. Kidney Int. 2006;70(4): Nephritis Trial. Ann Rheum Dis. 2010;69(12):2083–9.
732–42. 37. Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ,
20. Grootscholten C, Bajema IM, Florquin S, Steenbergen EJ, Peutz- Wofsy D, et al. Mycophenolate versus azathioprine as mainte-
Kootstra CJ, Goldschmeding R, et al. Treatment with cyclo- nance therapy for lupus nephritis. N Engl J Med. 2011;365(20):
phosphamide delays the progression of chronic lesions more 1886–95.
effectively than does treatment with azathioprine plus methyl- 38. Bomback AS, Appel GB. Updates on the treatment of lupus
prednisolone in patients with proliferative lupus nephritis. nephritis. J Am Soc Nephrol. 2010;21(12):2028–35.
Arthritis Rheum. 2007;56(3):924–37. 39. Mok CC, Ying KY, Yim CW, Ng WL, Wong WS. Very long-
21. Arends S, Grootscholten C, Derksen RH, Berger SP, de Sevaux term outcome of pure lupus membranous nephropathy treated
RG, Voskuyl AE, et al. Long-term follow-up of a randomised with glucocorticoid and azathioprine. Lupus. 2009;18(12):
controlled trial of azathioprine/methylprednisolone versus 1091–5.
cyclophosphamide in patients with proliferative lupus nephritis. 40. Szeto CC, Kwan BC, Lai FM, Tam LS, Li EK, Chow KM, et al.
Ann Rheum Dis. 2011. Tacrolimus for the treatment of systemic lupus erythematosus
22. Isenberg D, Appel GB, Contreras G, Dooley MA, Ginzler EM, with pure class V nephritis. Rheumatology (Oxford). 2008;47(11):
Jayne D, et al. Influence of race/ethnicity on response to lupus 1678–81.
nephritis treatment: the ALMS study. Rheumatology (Oxford). 41. Tse KC, Lam MF, Tang SC, Tang CS, Chan TM. A pilot study on
2010;49(1):128–40. tacrolimus treatment in membranous or quiescent lupus nephritis
23. Ramos-Casals M, Diaz-Lagares C, Soto-Cardenas MJ, Brito- with proteinuria resistant to angiotensin inhibition or blockade.
Zeron P, Cuadrado MJ, Sanna G, et al. Rituximab therapy in Lupus. 2007;16(1):46–51.
lupus nephritis: current clinical evidence. Clin Rev Allergy 42. Kasitanon N, Petri M, Haas M, Magder LS, Fine DM. Myco-
Immunol. 2009;40(3):159–69. phenolate mofetil as the primary treatment of membranous lupus
24. Diaz-Lagares C, Croca S, Sangle S, Vital EM, Catapano F, nephritis with and without concurrent proliferative disease: a
Martinez-Berriotxoa A, et al. Efficacy of rituximab in 164 retrospective study of 29 cases. Lupus. 2008;17(1):40–5.
patients with biopsy-proven lupus nephritis: pooled data from 43. Radhakrishnan J, Moutzouris DA, Ginzler EM, Solomons N,
European cohorts. Autoimmun Rev. 2011;11:357–64. Siempos II, Appel GB. Mycophenolate mofetil and intravenous
25. Rea Furie. Efficacy and safety of rituximab in subjects with active cyclophosphamide are similar as induction therapy for class V
proliferative lupus nephritis (LN): results from the randomized, lupus nephritis. Kidney Int. 2010;77(2):152–60.
double-blind phase III LUNAR study [abstract]. Arthritis Rheum. 44. Dussol B, Morange S, Burtey S, Indreies M, Cassuto E, Mourad
2009;60[Suppl. 10]:a1149. G, et al. Mycophenolate mofetil monotherapy in membranous
26. Lee YH, Lee HS, Choi SJ, Dai Ji J, Song GG. Efficacy and safety nephropathy: a 1-year randomized controlled trial. Am J Kidney
of tacrolimus therapy for lupus nephritis: a systematic review of Dis. 2008;52(4):699–705.
clinical trials. Lupus. 2011;20(6):636–40. 45. Jonsdottir T, Sundelin B, Welin Henriksson E, van Vollenhoven
27. Miyasaka N, Kawai S, Hashimoto H. Efficacy and safety of RF, Gunnarsson I. Rituximab-treated membranous lupus nephri-
tacrolimus for lupus nephritis: a placebo-controlled double-blind tis: clinical outcome and effects on electron dense deposits. Ann
multicenter study. Mod Rheumatol. 2009;19(6):606–15. Rheum Dis. 2011;70(6):1172–3.
28. Chen W, Tang X, Liu Q, Fu P, Liu F, Liao Y, et al. Short-term 46. Fornoni A, Sageshima J, Wei C, Merscher-Gomez S, Aguillon-
outcomes of induction therapy with tacrolimus versus cyclo- Prada R, Jauregui AN, et al. Rituximab targets podocytes in
phosphamide for active lupus nephritis: a multicenter randomized recurrent focal segmental glomerulosclerosis. Sci Transl Med.
clinical trial. Am J Kidney Dis. 2011;57(2):235–44. 2011;3(85):85ra46.
29. Li X, Ren H, Zhang Q, Zhang W, Wu X, Xu Y, et al. Myco- 47. Dooley MA, Houssiau F, Aranow C, D’Cruz DP, Askanase Anca
phenolate mofetil or tacrolimus compared with intravenous D, Roth D, et al. Effect of belimumab treatment on renal out-
cyclophosphamide in the induction treatment for active lupus comes: results from phase 3 belimumab clinical trials in patients
nephritis. Nephrol Dial Transpl. 2012;27:1467–72. with systemic lupus erythematosus [abstract]. Arthritis Rheum.
30. Najafi CC, Korbet SM, Lewis EJ, Schwartz MM, Reichlin M, 2011;63[Suppl 10]:2472.
Evans J. Significance of histologic patterns of glomerular injury 48. Daikh DI, Wofsy D. Cutting edge: reversal of murine lupus
upon long-term prognosis in severe lupus glomerulonephritis. nephritis with CTLA4Ig and cyclophosphamide. J Immunol.
Kidney Int. 2001;59(6):2156–63. 2001;166(5):2913–6.
31. Bao H, Liu ZH, Xie HL, Hu WX, Zhang HT, Li LS. Successful 49. Merrill JT, Burgos-Vargas R, Westhovens R, Chalmers A,
treatment of class V ? IV lupus nephritis with multitarget ther- D’Cruz D, Wallace DJ, et al. The efficacy and safety of abatacept
apy. J Am Soc Nephrol. 2008;19(10):2001–10. in patients with non-life-threatening manifestations of systemic
32. Lanata CM, Mahmood T, Fine DM, Petri M. Combination ther- lupus erythematosus: results of a twelve-month, multicenter,
apy of mycophenolate mofetil and tacrolimus in lupus nephritis. exploratory, phase IIb, randomized, double-blind, placebo-con-
Lupus. 2010;19(8):935–40. trolled trial. Arthritis Rheum. 2010;62(10):3077–87.
33. Rihova Z, Vankova Z, Maixnerova D, Dostal C, Jancova E, Honsova 50. Furie R, Nicholls K, Cheng T-T, Houssiau F, Burgos-Vargas R,
E, et al. Treatment of lupus nephritis with cyclosporine—an outcome Chen S-L, et al. Efficacy and safety of abatacept over 12 months
analysis. Kidney Blood Press Res. 2007;30(2):124–8. in patients with lupus nephritis: results from a multicenter, ran-
34. Zavada J, Pesickova S, Rysava R, Olejarova M, Horak P, Hrncir domized, double-blind, placebo-controlled phase II/III study
Z, et al. Cyclosporine A or intravenous cyclophosphamide for [abstract]. Arthritis Rheum. 2011;63[Suppl 10]:2469.
lupus nephritis: the Cyclofa-Lune study. Lupus. 2010;19(11): 51. Wofsy D, Shropshire Stephanie M, Hillson JL, Diamond B.
1281–9. Abatacept for lupus nephritis: alternative outcome measures
35. Contreras G, Pardo V, Leclercq B, Lenz O, Tozman E, O’Nan P, support opposing interpretations of data from a multicenter,
et al. Sequential therapies for proliferative lupus nephritis. N Engl randomized, double-blind, placebo-controlled phase II/III study
J Med. 2004;350(10):971–80. [abstract]. Arthritis Rheum. 2011;63[Suppl 10]:2474.

123
Mod Rheumatol

52. Westerweel PE, Luyten RK, Koomans HA, Derksen RH, Verhaar The Canadian Hydroxychloroquine Study Group. Lupus.
MC. Premature atherosclerotic cardiovascular disease in systemic 1998;7(2):80–5.
lupus erythematosus. Arthritis Rheum. 2007;56(5):1384–96. 61. Pons-Estel GJ, Alarcon GS, McGwin G Jr, Danila MI, Zhang J,
53. Kahlenberg JM, Kaplan MJ. The interplay of inflammation and Bastian HM, et al. Protective effect of hydroxychloroquine on
cardiovascular disease in systemic lupus erythematosus. Arthritis renal damage in patients with lupus nephritis: LXV, data from a
Res Ther. 2011;13(1):203. multiethnic US cohort. Arthritis Rheum. 2009;61(6):830–9.
54. Alarcon GS, Calvo-Alen J, McGwin G, Jr, Uribe AG, Toloza SM, 62. Kasitanon N, Fine DM, Haas M, Magder LS, Petri M. Hydrox-
Roseman JM, et al. Systemic lupus erythematosus in a multi- ychloroquine use predicts complete renal remission within
ethnic cohort: LUMINA XXXV. Predictive factors of high dis- 12 months among patients treated with mycophenolate mofetil
ease activity over time. Ann Rheum Dis. 2006;65(9):1168–74. therapy for membranous lupus nephritis. Lupus. 2006;15(6):
55. Duran-Barragan S, McGwin G Jr, Vila LM, Reveille JD, Alarcon 366–70.
GS. Angiotensin-converting enzyme inhibitors delay the occur- 63. Ruiz-Irastorza G, Ramos-Casals M, Brito-Zeron P, Khamashta
rence of renal involvement and are associated with a decreased MA. Clinical efficacy and side effects of antimalarials in systemic
risk of disease activity in patients with systemic lupus erythe- lupus erythematosus: a systematic review. Ann Rheum Dis.
matosus—results from LUMINA (LIX): a multiethnic US cohort. 2010;69(1):20–8.
Rheumatology (Oxford). 2008;47(7):1093–6. 64. Faurschou M, Dreyer L, Kamper AL, Starklint H, Jacobsen S.
56. Norby GE, Holme I, Fellstrom B, Jardine A, Cole E, Abedini S, Long-term mortality and renal outcome in a cohort of 100
et al. Effect of fluvastatin on cardiac outcomes in kidney trans- patients with lupus nephritis. Arthritis Care Res (Hoboken). 2010;
plant patients with systemic lupus erythematosus: a randomized 62(6):873–80.
placebo-controlled study. Arthritis Rheum. 2009;60(4):1060–4. 65. Ayodele OE, Okpechi IG, Swanepoel CR. Predictors of poor
57. Aprahamian T, Bonegio R, Rizzo J, Perlman H, Lefer DJ, Rifkin renal outcome in patients with biopsy-proven lupus nephritis.
IR, et al. Simvastatin treatment ameliorates autoimmune disease Nephrology (Carlton). 2010;15(4):482–90.
associated with accelerated atherosclerosis in a murine lupus 66. Weng CH, Hsu CW, Yu CC, Yen TH, Yang CW, Hung CC.
model. J Immunol. 2006;177(5):3028–34. Peritoneal dialysis and hemodialysis in systemic lupus erythe-
58. Shimazu H, Kinoshita K, Hino S, Yano T, Kishimoto K, Nagare matosus patients: comparison of clinical outcomes. Kidney Blood
Y, et al. Effect of combining ACE inhibitor and statin in lupus- Press Res. 2009;32(6):451–6.
prone mice. Clin Immunol. 2010;136(2):188–96. 67. Rietveld A, Berden JH. Renal replacement therapy in lupus
59. Lee SJ, Silverman E, Bargman JM. The role of antimalarial nephritis. Nephrol Dial Transpl. 2008;23(10):3056–60.
agents in the treatment of SLE and lupus nephritis. Nat Rev 68. Kang SH, Chung BH, Choi SR, Lee JY, Park HS, Sun IO, et al.
Nephrol. 2011;7(12):718–29. Comparison of clinical outcomes by different renal replacement
60. Tsakonas E, Joseph L, Esdaile JM, Choquette D, Senecal JL, therapy in patients with end-stage renal disease secondary to
Cividino A, et al. A long-term study of hydroxychloroquine lupus nephritis. Korean J Intern Med. 2011;26(1):60–7.
withdrawal on exacerbations in systemic lupus erythematosus.

123

View publication stats

You might also like