Sunteți pe pagina 1din 21

Autoimmunity Reviews 12 (2012) 174–194

Contents lists available at SciVerse ScienceDirect

Autoimmunity Reviews
journal homepage: www.elsevier.com/locate/autrev

Review

Lupus nephritis: A critical review


Andrea T. Borchers a, Naama Leibushor a, Stanley M. Naguwa a, Gurtej S. Cheema a,
Yehuda Shoenfeld b, M. Eric Gershwin a,⁎
a
Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA 95616, United States
b
Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center (affiliated to Tel-Aviv University), Tel-Hashomer 52621, Israel

a r t i c l e i n f o a b s t r a c t

Article history: Lupus nephritis remains one of the most severe manifestations of systemic lupus erythematosus associated
Accepted 12 August 2012 with considerable morbidity and mortality. A better understanding of the pathogenesis of lupus nephritis is
Available online 8 September 2012 an important step in identifying more targeted and less toxic therapeutic approaches. Substantial research
has helped define the pathogenetic mechanisms of renal manifestations and, in particular, the complex role
Keywords:
of type I interferons is increasingly recognized; new insights have been gained into the contribution of
Tolerance
Immune complexes
immune complexes containing endogenous RNA and DNA in triggering the production of type I interferons
Interferons by dendritic cells via activation of endosomal toll-like receptors. At the same time, there have been
Autoantibodies considerable advances in the treatment of lupus nephritis. Corticosteroids have long been the cornerstone
of therapy, and the addition of cyclophosphamide has contributed to renal function preservation in patients
with severe proliferative glomerulonephritis, though at the cost of serious adverse events. More recently, in
an effort to minimize drug toxicity and achieve equal effectiveness, other immunosuppressive agents,
including mycophenolate mofetil, have been introduced. Herein, we provide a detailed review of the trials
that established the equivalency of these agents in the induction and/or maintenance therapy of lupus
nephritis, culminating in the recent publication of new treatment guidelines by the American College of
Rheumatology. Although newer biologics have been approved and continue to be a focus of research,
they have, for the most part, been relatively disappointing compared to the effectiveness of biologics in
other autoimmune diseases. Early diagnosis and treatment are essential for renal preservation.
© 2012 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
2. Histological classification of lupus nephritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
3. Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176
3.1. Breaking of tolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176
3.2. Pathogenesis of LN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
3.3. Amplification mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
3.3.1. The type I interferon axis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
3.3.2. T cells in lupus nephritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
3.3.3. IL-17 producing T cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
3.4. Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
4. Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
4.1. Treatment of class I and II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
4.2. Treatment of class III and IV . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
4.2.1. The NIH and Euro-Lupus (ELNT) regimens . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
4.2.2. Mycophenolate mofetil (MMF) as induction therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
4.2.3. AZA plus methylprednisolone for induction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183
4.2.4. MMF or AZA for maintenance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184

⁎ Corresponding author at: Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite
6510, Davis, CA 95616, United States. Tel.: +1 530 752 2884; fax: +1 530 752 4669.
E-mail address: megershwin@ucdavis.edu (M.E. Gershwin).

1568-9972/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.autrev.2012.08.018
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 175

4.3. Treatment of membranous LN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184


4.4. Biological agents as further treatment options . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
4.4.1. Belimumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
4.4.2. Rituximab and ocrelizumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
4.4.3. Abatacept and other biologicals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
5. Biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
6. Outcome/prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
6.1. Remission and flares . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
6.2. Progression to ESRD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186
6.3. Renal replacement therapy treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186
6.4. Patient survival . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187
6.5. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 187
Take-home messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 188

1. Introduction only about 22% of the variability in the occurrence of renal involve-
ment. In several cohort studies, male SLE patients were shown to
Systemic lupus erythematosus (SLE) is an autoimmune disease have more renal involvement and worse outcome compared to their
that can affect essentially any organ or tissue. It is the prototypic female counterparts [37–40]. However, this gender disparity was
autoimmune disease [1,2]. One of its most severe manifestations is not shown in other cohorts [41,42]. We should also note that much
lupus nephritis (LN), which remains a cause of substantial morbidity of our knowledge or mechanism is based on murine models [43–51].
and mortality, either secondary to kidney disease, or as a result of
intense immunosuppressive drug toxicity [3–10]. The incidence and 2. Histological classification of lupus nephritis
prevalence of SLE overall generally range from 1 to 5/10 5 and 20 to
150/10 5, respectively, with women being affected 9 times more Glomerulonephritis (GN) is the most common form of renal
frequently than men [11]. The frequency of SLE is 2- to 8-fold higher disease in patients with SLE, but is frequently accompanied by
in non-European populations, particularly those of African ancestry. tubulointerstitial and/or vascular lesions (such as thrombosis second-
LN epidemiology data are limited. One study from northwest ary to antiphospholipid syndrome). A variety of other nephropathies,
England yielded incidence and prevalence of 0.4/10 5 and 4.4/10 5, including renal amyloidosis, focal segmental glomerulosclerosis,
respectively [12]. The onset of SLE in most populations occurs most minimal-change disease, IgA and IgM nephropathy, necrotizing
commonly during the third and fourth decade of life and somewhat glomerulitis, sarcoidal and NSAID-induced tubulointerstitial nephri-
later in populations of European descent. Approximately 15–20% of tis, and thin basement membrane disease have also been described
patients experience the onset of SLE prior to 18 years of age. The in patients with SLE [52]. Lupus nephritis can present as nephritic
frequency of renal involvement is particularly high in juvenile-onset and/or nephrotic syndrome with various combinations of edema,
SLE, ranging from 50% to 80% in most cohorts described to date constitutional symptoms, proteinuria, hematuria, impaired renal
[13–18] and does not differ with ethnicity [14]. In contrast, LN is function, abnormal lipid profile and hypertension. The clinical pre-
less frequent in patients with late-onset SLE (≥50 years), with sentation does not correlate very well with the type and severity of
fewer than 30% of patients affected [19]. Renal disease may be the renal biopsy histology findings. Hence, kidney biopsies remain the
first manifestation of SLE, but most commonly occurs within a year gold standard for establishing the diagnosis of LN, for determining
of diagnosis, and almost always within 5 years, but can occur any the activity and chronicity of the disease and formulating the appro-
time throughout the course of the disease [20,21]. Age-standardized priate type of treatment.
prevalence differs significantly by ethnicity, being 3.5/10 5 for white, The histological changes found in biopsy samples of LN kidneys
13.3 for Indo-Asian, 64.6 for Afro-Caribbean and 66.7 for Chinese are highly diverse, ranging from mild mesangial involvement,
patients. This is in agreement with numerous cohort studies demon- through proliferative forms of GN to end stage fibrosis, including
strating that SLE patients of European descent have the lowest fre- endothelial injury, endocapillary proliferation and membranous LN
quency of renal disease (20–45%) [20,22–24], whereas 50–70% of characterized by podocyte injury and nonproliferative capillary wall
SLE patients develop nephritis in African American and certain lesion. Importantly, various combinations of these lesions can co-
Asian, Arab, Hispanic, indigenous and mestizo populations [11]. exist. Consequently, there have been several attempts to classify LN
Non-European patients, in particular those with African ancestry, biopsies according to the most prominent pattern of histologic
often develop LN earlier in the course of the disease [21,25–29]. abnormality, first by the WHO and more recently by the International
African ancestry has been identified as an independent predictor Society of Nephrology and the Renal Pathology Society (ISN/RPS)
of developing renal disease at any time during the disease course, [53,54] (see Table 1). Both classification systems are exclusively
particularly in close proximity to SLE diagnosis in various multiethnic concerned with the glomerular pathology; tubulointerstitial and
cohorts [21,26–28]. Texan Hispanic ethnicity independently predict- vascular lesions are not addressed, although the ISN/RPS classification
ed the development of LN in close proximity to SLE diagnosis, but contains a strong recommendation to report such lesions. The
not renal involvement in general [27,28]. A variety of other demo- most important changes in the ISN/RPS compared to the WHO
graphic, serological, clinical and genetic factors have also been classification include: 1) class I no longer includes a “Nil” category,
implicated [21,26–28,30], and education [28] and socioeco- i.e., the complete lack of renal abnormalities by light microscopy,
nomic variables were identified as risk factors for developing immunofluorescence, and electron microscopy is excluded from the
LN [21,26,31–35]; the “ethnicity” variable itself was found to be ISN/RPS classification system; 2) qualitative and quantitative differ-
explained in part by admixture, particularly of African American an- ences are introduced into the distinction between class III and class
cestral genes, and in part by a composite measure of socioeconomic IV lesions; 3) class IV is subdivided into a global and a segmental sub-
status, but at least 45% of this variable remained unexplained [36]. group; 4) rather than subdividing class V into subgroups depending
Overall, ethnicity and socioeconomic status combined accounted for on the presence or absence of class II, III, or IV lesions, the use of
176 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

Table 1
WHO and ISN/RPS classifications of lupus nephritis (LN).

WHO ISN/RPS

Class Normal glomeruli Minimal mesangial LN


I a. Nil (by all techniques) Normal glomeruli by LM, but mesangial immune deposits by IF
b. Normal by light microscopy, but deposits by IF or electron microscopy
Class Pure mesangial alterations Mesangial proliferative LN
II a. Mesangial widening and/or mild hypercellularity (+) Pure mesangial hypercellularity of any degree or mesangial matrix expansion by light
microscopy, with mesangial immune deposits
b. Moderate hypercellularity (++) Maybe a few isolated subepithelial or subendothelial deposits visible by IF or electron
microscopy, but not by LM
Class Focal segmental glomerulonephritis Focal LN
III Active or inactive focal, segmental or global GN involving b50% of all glomeruli
a. With active necrotizing lesions Class III (A) active lesions: focal proliferative LN
b. With active and sclerosing lesions Class III (A/C) active and chronic lesions: focal proliferative and sclerosing LN
c. With sclerosing lesions Class III (C) chronic inactive lesions with glomerular scars: focal sclerosing LN
Class Diffuse glomerulonephritis (severe mesangial, endocapillary or Diffuse LN
IV mesangiocapillary proliferation and/or extensive subendothelial deposits)
a. Without segmental lesions Active or inactive diffuse, segmental or global GN involving ≥50% of all glomeruli; divided into:
– diffuse segmental (IV-S) LN when ≥50% of the involved glomeruli have segmental
lesions (involving less than half of the glomerular tuft);
– diffuse global (IV-G) LN when ≥50% of the involved glomeruli have global lesions.
b. With active necrotizing lesions Class IV-S (A) active lesions: diffuse segmental proliferative LN
Class IV-G (A) active lesions: diffuse global proliferative LN
c. With active and sclerosing lesions Class IV-S (A/C) active and chronic lesions: diffuse segmental proliferative and sclerosing
LN
Class IV-G (A/C) active and chronic lesions: diffuse global proliferative and sclerosing LN
d. With sclerosing lesions Class IV-S (C) chronic inactive lesions with scars: diffuse segmental sclerosing LN
Class IV-G (C) chronic inactive lesions with scars: diffuse global sclerosing LN
Class Diffuse membranous glomerulonephritis Membranous LN
V a. Pure membranous glomerulonephritis Global or segmental subepithelial immune deposits or their morphologic sequelae by LM
and by IF or electron microscopy, with or without mesangial alterations
b. Associated with lesions of class II Class V LN may occur in combination with class III or IV, in which case both will be diagnosed
c. Associated with lesions of class III Class V LN demonstrates advanced sclerosis
d. Associated with lesions of class IV
Class Advanced sclerosing glomerulonephritis Advanced sclerotic LN
VI ≥90% of glomeruli globally sclerosed without residual activity

GN = glomerulonephritis; IF = immunofluorescence; LM = light microscopy.


WHO = World Health Organization.
ISN/RPS = International Society of Nephrology and the Renal Pathology Society.

combined designations is encouraged in the ISN/RPS classification previously classified as WHO class III ≥50% were switched to ISN/RPS
system (e.g., class III and class V or class IV and class V). class IV-S, the remainder were classified as IV-G [64]. This clearly
The most controversial of these changes in the ISN/RPS classification demonstrates that WHO III ≥50% and class IV differ from ISN/RPS
system is the subdivision of class IV into diffuse global and diffuse seg- classes IV-S and IV-G, respectively. The authors suggest that important
mental LN. This was based largely on the results of a single investigation clinical and prognostic information is lost by adopting the ISN/RPS
by Najafi et al. [55] demonstrating that patients with focal segmental class IV-G and IV-S categories. This may explain why none of the analy-
GN with ≥50% of glomeruli involvement (WHO class III ≥50%) had sig- ses reported to date has been able to demonstrate a significant differ-
nificantly worse 5- and 10-year renal survival rates compared to pa- ence in prognosis or in outcome between patients with ISN/RPS class
tients with WHO class IV, Vc or Vd lesions, and were less likely to IV-G and IV-S lesions at an initial biopsy [57,58,60–63]. However, global
enter remission compared to class IV. Several studies examined wheth- lesions in a second biopsy obtained after therapy may have prognostic
er the subclassification of class IV LN into segmental (involving less than value since all of the patients with global lesions experienced doubling
half of the glomeruli) versus global (involving more than half of the glo- of serum creatinine, but only 36% of those with segmental lesions
meruli) truly reflects clinical differences or adds prognostic value experienced doubling of serum creatinine. However, this difference
[56–63]. Class IV-global (class IV-G) was associated with more signifi- did not reach statistical significance due to the small number of subjects
cant proteinuria and nephrotic syndrome, higher serum creatinine, (n=17) [57]. Most of these studies using the ISN/RPS classification
hypertension, and more pronounced hypocomplementemia, although system did not subdivide class IV lesions into active (A), active and
these differences were not always consistent, and did not always chronic (A/C) and chronic (C). Activity/chronicity score seems to be of
reach statistical significance. Generally, other typical histologic findings high prognostic value as Japanese patients with LN class IV-G (A)
of class IV-G lesions were endocapillary proliferation and wire loops, responded well to therapy, and none experienced doubling of serum
whereas fibrinoid necrosis was more frequent in class IV-S defused creatinine, compared to those with LN class IV (A/C), whose renal
segmental lesions [57,58,62,63]. A variety of other morphological outcome was significantly worse [60].
features have also been found to differ significantly between classes
IV-S and IV-G in individual studies, but the overall results are highly 3. Pathogenesis
inconsistent. This is partly due to high interobserver variability for clas-
sifying renal biopsy specimens, regardless of whether the WHO 1995 or 3.1. Breaking of tolerance
the ISN/RPS system was used. In one study where interobserver agree-
ment was scored, only 15% of 54 individual histological characteristics SLE is characterized by the production of numerous autoanti-
were scored as “good”, whereas 54% of these items reached “poor” [63]. bodies, which predominantly target nuclear antigens, and these are
Interestingly, when the ISN/RPS classification system was applied to strongly implicated in the pathogenesis of LN [65]. Although the rea-
the biopsies of the patients studied by Najafi et al. [55], only half of those son for the targeting of nuclear antigens is still not fully understood,
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 177

some evidence suggest that it relates to the expression of lupus Renal immune deposits are found in virtually all patients with SLE,
autoantigens on the surface of apoptotic blebs [66], increased apopto- yet a substantial proportion of them retain normal histology by light
sis, and the defective clearance of apoptotic bodies [67,68]. There are microscopy [83,84]. Other patients have histological abnormalities,
indications that apoptotic nucleosomes, i.e., chromatin, are the initial frequently representing proliferative or membranous glomerulone-
immunogen in SLE, with subsequent spreading of the reactivity to phritis, but never developing clinical manifestations of renal disease
histones and dsDNA [69]. However, there are patients with anti- [84–86]. Such cases of “silent nephritis” indicate that the progression
dsDNA antibodies who do not display detectable levels of anti- from immune deposits to morphological alterations, and to the devel-
chromatin reactivity, hence, there are probably other mechanisms opment of clinical symptoms requires other events or factors. Those
leading to anti-dsDNA reactivity. must play a role in either enhancing susceptibility to, or protecting
Usually apoptotic bodies are cleared rapidly from the circulation from these processes. This is further supported by studies in certain
and undergo secondary necrosis if clearance is deficient. It has been murine models where experimental animals fail to develop inflam-
shown that a subset of nucleosomes released from cells in the late matory renal disease despite marked deposition of ICs [78,87] or
stages of apoptosis, i.e., secondary necrosis, contains high mobility where clinical disease does not develop despite considerable glomer-
group box protein 1 (HMGB1). This is a protein found only in SLE ular pathology [88].
patients' sera that can both stabilize nucleosome structure and Although more than 150 different antigens can be targeted in SLE, nu-
turn into a proinflammatory molecule once it is released from cells clear antigens are most strongly implicated in the pathogenesis of LN.
[70]. HMGB1 nucleosome complexes are capable of inducing These include antibodies against DNA (both double stranded and single
dendritic cell (DC) maturation and of stimulating the release of pro- stranded), chromatin, histone, SSA, SSB, and ribonucleoprotein. All of
inflammatory cytokines from macrophages in a TLR-2-dependent these as well as anti-C1q antibodies have been eluted from postmortem
manner [70]. Immunization of mice with late apoptotic nucleosomes kidney tissues of patients with LN, and were found in higher serum quan-
induces the production of antibodies to dsDNA, to histones and – in tities compared to surrogate (the initial supernatant of glomeruli)
some mice – to extractable nuclear antigens, again in a TLR-2- [89–94]. Of note, enrichment of anti-histone and anti-chromatin reactiv-
dependent manner. This suggests that HMGB1-nucleosome com- ity was markedly more frequent compared to enrichment of anti-double
plexes may be a crucial factor in breaking tolerance to nuclear anti- stranded DNA antibodies [89]. This supports the hypothesis that chroma-
gens. Patients with SLE exhibit both elevated serum HMGB1 as well tin constitutes the initial target of autoreactivity in SLE.
as antibodies to HMGB1 [71–73]. An association of serum HMGB1 Anti-dsDNA antibodies are a hallmark of SLE and represent the most
levels with renal disease, particularly active renal disease, has been thoroughly investigated antibody specificity. They are detectable long
suggested [71,73], but a longitudinal study did not reveal significant before the development of SLE and LN [95] and are found more
reduction in serum HMGB1 concentrations during therapy [72]. frequently and at higher titers in patients with LN compared to SLE
HMGB1 was also found to be strongly expressed in renal biopsy tissue patients without renal involvement [96,97]. However, this is not an
from all WHO classes of LN, with intense immunostaining outlining entirely consistent finding, and the ability of anti-dsDNA antibodies to
the glomerular endothelium and to a lesser extent in the mesangium, predict renal flares remains controversial [98]. Anti-dsDNA antibodies
whereas control tissue demonstrated almost exclusively nuclear demonstrate evidence of somatic hypermutation, indicating that their
staining [72]. The precise role of renal target tissue expression of development is antigen driven. The most direct evidence for the patho-
HMGB1 remains to be determined. genicity of anti-dsDNA antibodies comes from the demonstration of its
production and subsequent renal deposition and proteinuria in severe
combined immunodeficiency (SCID) mice following intraperitoneal
3.2. Pathogenesis of LN hybridoma administration [99]. Despite great efforts it has not been
possible to fully define the characteristics which make certain anti-
While the pathogenesis of LN remains incompletely understood, dsDNA antibodies pathogenic versus inertic. Isotype certainly plays a
major insights are derived from a variety of animal models which cap- role, and IgG subclass also appears to be more pathogenic than IgA or
ture multiple features of SLE, and in particular glomerulonephritis. IgM, most likely because of differential affinities for complement and
These models include MRL/lpr, NZB/W F1 hybrid (NZB/W) mice, and FcγR. The antibody specificity has been implicated in determining the
the NZB/W-derived congenic strains like NZM 2328 and NZM 2410. localization of IC deposits as well as the subsequent pathological and
There is evidence from these animal models that glomerulonephritis clinical changes [100]. In addition, nephritogenic anti-dsDNA antibodies
is initiated by renal deposition or formation of immune complexes often display high avidity as well as cross-reactivity with a variety of
(ICs), resulting in the activation of complement and Fcγ receptors intracellular and extracellular antigens. Their charge may also be impor-
(FcγR). The complement system has a dual role in SLE and LN. Homozy- tant [96,101–104].
gous deficiency of C1 components and, to a lesser extent, C2 and C4 is How anti-dsDNA and other autoantibodies deposit in the kidney is
the strongest single genetic risk factor for the development of SLE, also not fully elucidated. The original theory that ICs in the circulation
most likely because of the complement's role in clearing apoptotic become passively trapped in glomeruli is not supported by evidence,
bodies and ICs [74]. Recently anti-C1q Abs specific to the globular and is now considered unlikely. It has been hypothesized that cross-
head region of the molecule have been described [75]. Some of these reactivity with certain extracellular matrix molecules of the glomerular
autoantibodies are able to inhibit the interactions of C1q with IgG and basement membrane, such as laminin, type IV collagen, and heparan
CRP, suggesting their contribution to functional C1q deficiency as well sulfate, determines the glomerular localization of anti-dsDNA antibodies
as to defective clearance of apoptotic materials and ICs as observed in [105]. Other cross-reactive targets include intracellular structural pro-
many patients with SLE and LN. On the other hand, the results of animal teins, such as α-actinin-4 and myosin [102,104–106]. Importantly,
studies clearly implicate complement activation, particularly of the there are data suggesting that mesangial cells of MRL/lpr mice express
alternative pathway, in glomerular injury [76,77]. Although there is significantly higher levels of surface α-actinin compared to those of
also clear evidence for an important role of FcγR engagement in the Balb/c mice [102]. This suggests that antigen availability may be a
development of glomerulonephritis [78–80], the exact contribution of major determinant of whether LN develops or not. Interestingly, immu-
the specific activating and inhibiting receptors to tissue injury has not nization of non-autoimmune BALB/c mice with α-actinin resulted in
yet been fully elucidated. Another crucial step in the activation of high titers of antinuclear antibodies (ANA) that cross-reacted with chro-
immune pathways that amplify the pathogenic mechanisms in LN is matin, histones, and Sm/RNP, but not with dsDNA, anti-single stranded
the costimulation of FcγRs and endosomal toll-like receptors (TLRs) DNA, or cardiolipin [107]. Mice immunized with α-actinin developed
[81,82] (as will be discussed in more detail below). higher levels of renal immuloglobulin (Ig) deposition and proteinuria
178 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

compared to adjuvant-immunized control mice. However, several stud- thereby increasing the production of autoantibodies. In addition, acti-
ies indicate that, in vivo, there is no direct binding of anti-dsDNA anti- vated mDCs can stimulate CD8+ T cells to differentiate into cytotoxic
bodies to glomerular basement membrane components in both effector T cells. By causing tissue damage, such cytotoxic T lymphocytes
experimental animal models and in SLE patients [108–110]. Instead, (CTL) generate autoantigens, in particular nucleosomes, that can then
these studies provide evidence that the binding of these autoantibodies be presented by mDCs to ultimately activate yet more autoreactive
to membrane structures is mediated by interactions between antibody- B cells. Several lines of evidence strongly implicate type I IFNs in the
bound nucleosomes and heparan sulfate. These nucleosomes may derive pathogenesis of SLE: 1) treatment of patients with viral diseases or
from the circulation, or may arise locally from increasing apoptosis of malignancies with exogenous IFNα can induce autoantibodies and
intraglomerular cells, secondary to established nephritis. lupus-like disease; 2) approximately half of SLE patients harbor elevat-
While it is generally assumed that breaking of tolerance to nuclear ed serum levels of INFα [128,129] or demonstrate a so-called “type I IFN
antigens is central to LN pathogenesis, there is clear evidence from gene signature”, i.e., increased expression of type I IFN-regulated genes
animal models that renal IC deposition, severe proteinuria and chron- in peripheral blood [130,131]. This upregulation is being attributable
ic glomerulonephritis can develop in the absence of ANA, anti-DNA almost exclusively to IFNα [132]; 3) serum from some SLE patients is
and anti-nucleosome antibodies [111,112]. This implies that auto- capable of inducing the differentiation of blood monocytes into DCs in
antibodies without nuclear specificity can be nephritogenic. A non- an IFNα-dependent manner [133]; 4) several studies, though not all,
nuclear antigen of special interest in LN is C1q. In different studies, demonstrate an association between a type I IFN gene signature and
antibodies to C1q, which recognize epitopes on the collagen-like disease activity [134–136], although in longitudinal studies, changes
region of this complement component, are found at highly variable in disease activity do not necessarily correlate to the expression level
portions in SLE patients (ranging from 17 to 70%, depending of IFN-inducible genes [137,138].
partly on the assay system and the cut-off used for positivity) Data on an association of type I IFN gene significance with LN,
[97,113–116]. Serum C1q antibody positivity strongly correlates particularly active renal disease, are somewhat more controversial
with renal disease, with higher prevalence and titers in patients [130,134–136], with the largest study to date not detecting any asso-
with LN compared to those without, and in patients with active ciation [139]. Note that this study used a reporter assay to measure
renal disease compared to those with inactive renal disease serum IFNα activity. In contrast, the type I IFN signature or IFNα
[97,113,114,116–119]. Rising titers were found to predict renal flares score may actually reflect not only increased IFNα activity but also
better than did anti-dsDNA antibodies, particularly in patients with raise individual sensitivity to this cytokine [140]. Importantly, it has
proliferative GN [120,121], whereas anti-C1q reactivity decreased or recently been reported that resident renal cells can be a major source
even disappeared during successful therapy [117,119,121]. Several of IFNα in a murine model of immune-mediated nephritis and are,
groups of investigators found that lack of anti-C1q antibodies had therefore, likely to contribute to end-organ disease [141].
up to 100% negative predictive value [117,118,122,123], suggesting In various murine models of SLE or SLE phenotypes, deficiency of
that active LN, particularly class III or IV, does not occur in the absence type I IFN receptor (IFN-RI) markedly attenuates autoantibody pro-
of this reactivity. Lack of assay standardization and the failure to duction and reduces the frequency of renal disease [142–144]. This
obtain serum samples at the time of the biopsy may explain is with the notable exception of MRL/lpr mice, in which IFN-RI defi-
the lower negative predictive values found in other studies ciency induces autoantibody synthesis and worsens renal pathology,
[114,116,124]. There are data from a non-autoimmune experimental suggesting a role for type I IFN in suppressing B cell activation in
model suggesting that anti-C1q autoantibodies do not capture circu- this particular model [145]. Conversely, administration of adenovirus
lating C1q, but instead bind to the C1q present in ICs deposited in expressing IFNα results in prolonged production of this cytokine, en-
glomeruli, resulting in local complement and FcγR activation [125]. hances autoantibody production and accelerates the onset of protein-
This could explain why most patients demonstrate glomerular Ig uria and glomerulonephritis in the NZB/W and related mice models of
and C1q deposits, but overt renal disease develops only in those lupus [146–148]. It also renders these mice as more resistant to treat-
who also express anti-C1q antibodies. Direct binding of anti-C1q anti- ment that usually prevents the onset of proteinuria in conventional
bodies to nucleosomes and glomerular endothelial cells with subse- NZB/W mice and is associated with a higher relapse rate after
quent enhancement of C3 deposition, particularly in the presence of remission-inducing therapy of established kidney disease [149].
anti-DNA antibodies, may also result in increased complement activa- The major producers of IFNα are plasmacytoid DCs (pDCs). Evidence
tion and glomerular injury [126]. However, patients with LN without has been accumulating that endogenous RNA or DNA in the form of ICs
anti-C1q antibodies at the time of biopsy have been described, includ- can induce IFNα production in the pDCs of healthy volunteers
ing up to one-third of patients with class III and IV lesions [127], and [82,150–152]. RNA-containing ICs signal through Toll like receptor
renal flares can also develop in the absence of detectable anti-C1q (TLR)7, whereas DNA containing ICs signal through TLR9 [151,153].
antibodies [121]. Both TLRs are endosomal receptors, and FcγRIIa was shown to be the
The sum of the seven identified antibody specificities found to be cell membrane receptor that delivers DNA-containing ICs to the intra-
enriched in kidney eluates of patients with LN accounts for less than cellular lysosomes containing TLR9 [82]. There are data strongly
10% of the total eluted IgG in most patients (the range being b 1% to suggesting that IC-induced secretion of IFNα by pDCs depends on the
a maximum of 41%) [89]. This suggests that a variety of other autoan- presence of HMGB1 in these complexes, and requires the HMGB1 re-
tibody specificities are deposited in lupus kidneys, and may partici- ceptor, RAGE [153]. Interestingly, C1q can inhibit IC-induced production
pate in renal pathology. of IFNα by pDCs. However, it remains unclear whether this is due to a
direct effect of C1q on pDCs [154], or occurs via an indirect mechanism
3.3. Amplification mechanisms involving preferential binding of C1q-ICs to macrophages [155]. In
either case, these studies reveal another role for C1q in SLE pathophys-
3.3.1. The type I interferon axis iology and emphasizes that the functional C1q deficiency frequently ob-
Type 1 interferons (IFNs) are crucial mediators of anti-viral immune served in SLE patients may contribute to the continuous production of
responses and are important regulators of the proliferation, differentia- IFNα.
tion, survival and function of almost every immune cell type. Yet, they ICs containing chromatin or nucleosomes are also able to activate
also play an important role in autoimmune diseases, including SLE. murine mDCs, resulting in a distinct cytokine pattern consisting of in-
IFNα in particular can induce the differentiation of monocytes into creased TNFα production in the absence of IL-12 induction [81]. Such
DCs and can activate immature myeloid DCs (mDCs). This in turn acti- ICs also induce production of B cell activating factor (BAFF), which
vates autoreactive T cells and provides help to autoreactive B cells, plays an important role in B cell proliferation and activation. Whereas
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 179

cytokine induction was found to depend mostly on a pathway involv- B-cell aggregates with increasing levels of organization. The highest
ing FcγRIII and TLR9, the induction of BAFF was TLR9-independent level of organization consists of germinal centers with separate B
and could therefore be an important mechanism in humans, whose and T cell compartments around a central network of follicular DCs
mDCs do not express TLR9 [156]. [170,171]. Many of these B cells display a mature non-antibody se-
Another mechanism for activation of pDC by endogenous DNA in- creting phenotype with strong MHC class II expression, suggesting
volves neutrophil extracellular traps (NETs). These are web-like struc- that they are capable of antigen presentation [170]. This makes it like-
tures that arise from a specific type of cell death called NETosis and ly that germinal centers contribute to the persistence of renal inflam-
consist mainly of chromatin and neutrophil proteins. Interestingly, mation. Scattered plasma cells were detected in surrounding areas
anti-RNP antibodies induce NETosis in neutrophils of about two-thirds [170,171], raising the possibility of in situ autoantibody production,
of SLE patients, but not in those of healthy individuals, and this process as has been reported in NZB/W mice, although germinal centers
requires FcγRII and TLR7 [157]. Upon in vitro stimulation with the pro- were not detected in this model [172].
tein kinase C activator, phorbolmyristase acetate (PMA), neutrophils In LN kidney biopsies the mononuclear cell infiltrate is mainly
from SLE patients released more NET-DNA compared to those from found in a periglomerular and peritubular interstitial distribution,
healthy controls [158]. A subset of SLE patients was also found to exhibit and consists primarily of CD3+ T cells, as well as some CD20 + B
defective degradation of NETs, and this defect was significantly associ- cells and macrophages [173–176]. Fewer leukocytes are found within
ated with the presence of renal disease [159]. Importantly, NETs are the glomerulus, where monocytes/macrophages constitute the major
capable of triggering pDC activation and IFNα production in a TLR9- cell type aggregate. The patterns of distribution are quite variable, as
dependent manner [157,158]. NET uptake by PDCs requires the pres- is the CD4 + and CD8 + T cell ratio and its contribution to the total
ence of the antimicrobial peptide LL37. This was further enhanced by CD3+ T cell infiltrate, with CD8 + T cells dominating in some sam-
the presence of human neutrophil peptides (HNPs) [158]. Anti-LL37 ples and CD4+ T cells in others [173–175,177]. Several studies
and anti-HNP antibodies were detected in 41 and 58% of SLE patients, found a correlation between the composition of the infiltrate and
respectively. These antibodies were found to enhance the FCγRII- the severity of tubulointerstitial inflammation as well as the kidney
mediated internalization of DNA-containing ICs. NETs also contain damage and the risk of renal failure [174,178,179]. The infiltrating T
HMGB1, another protein that has been shown to be essential for cells in LN exhibit a CD28 null memory-effector phenotype, as do uri-
IC-induced IFNα production by pDCs [153]. nary CD8 T cells of LN patients [177,180]. Conversely, the proportion
Stimulation through TLR7 and TLR9 was found to protect human of circulating effector-memory CD8 + T cells was decreased in SLE
pDCs and other TLR9 positive cell types (conventional DCs and B cells) patients, particularly those with active renal disease, compared to
from glucocorticoid-induced cell death. Inhibition of TLR7 and TLR9 in healthy controls. This suggests the migration of effector-memory
vitro and in vivo restored glucocorticoid sensitivity, and reduced the ex- CD8+ T cells from the peripheral compartment to the kidney [180].
pression of IFN-regulated genes [160]. This suggests that exaggerated In addition, kidney-infiltrating T cells from LN patients demonstrate
TLR7 or TLR9 signaling could be responsible for the decreased cortico- evidence of oligoclonal expansion [177,181,182], specifically CD8+
steroid responsiveness in SLE patients compared to patients with T lymphocytes [177]. Furthermore, certain expanded clonotypes
other inflammatory diseases. were detected in different anatomical regions of the kidney as well
Administration of TLR7- or TLR9-specific ligands to MRL/lpr mice as in the peripheral blood. Certain clones were found to persist and
accelerates glomerulonephritis [161–164]. Studies in mice deficient even expand in patient's biopsy samples taken years apart. In conclu-
for TLR7 or TLR9 have shown that these receptors play important sion, these findings strongly argue for an effector function of CD8+ T
roles in the activation not only of pDCs, but also in the activation of cells in the progression of kidney injury.
B cells, and in the determination of autoantibody specificities [112]. In the NZM2328 lupus mouse model, the transition from acute to
TLR7 was found to be essential for the production of antibodies chronic GN, and the onset of severe proteinuria correspond to the in-
against RNA-containing antigens, such as Sm and Sm-RNP, whereas filtration of T cells and CD11 + DCs into the glomeruli, accompanied
TLR9 is required for the production of anti-chromatin and anti- by oligoclonal expansion of T cells in the kidney draining lymph
dsDNA antibodies. However, these receptors had opposite effects on nodes [183]. In LN patients, a decrease in circulating immature pDCs
clinical phenotype, with TLR7-deficient mice demonstrating less and certain subsets of mDCs correlates with active renal disease and
severe renal disease, whereas the absence of TLR9 was associated is associated with a simultaneous increase of both DC subsets in the
with more severe glomerulonephritis. These and subsequent data kidney particularly of patients with class III and IV LN [184,185]. DC
suggest that TLR9 has a more complex regulatory role via cross- accumulation was observed mostly in the glomeruli in one study
regulation of TLR7-dependent processes [165], and mandate some [185], but was more pronounced in the tubulointerstitium in another
caution in developing TLR-targeted therapeutic approaches. [184]. This suggests that DC-induced T cell activation may play a sim-
Glomerular expression of TLR9 has been detected in the biopsy ilar role in driving the progression of renal disease in LN patients.
samples of some patients with LN, but not in those of controls
[166,167]. There is some disagreement over whether tubulo- 3.3.3. IL-17 producing T cells
interstitial TLR9 expression is enhanced in LN samples compared to Of the IL-17 group of cytokines, IL-17A and IL-17F are best charac-
controls [166–168]. However, sera from SLE patients with anti- terized, and are known to play a central role in adaptive immune re-
dsDNA reactivity induced the expression of TLR9 in tubular epithelial sponses to bacteria and fungi. There is also growing evidence of their
cells and tubulointerstitial TLR9 expression was found to correlate implication in the pathogenesis of various autoimmune diseases, like-
with tubulointerstitial injury [168]. While this suggests an active ly because of their ability to induce pro-inflammatory responses
role of tubular and possibly glomerular cells in renal inflammation [186]. It is generally thought that the major cellular source of these
and tissue damage, the precise mechanisms remain to be elucidated. cytokines is Th17 cells. T cells producing IL-17 are increased in
the peripheral blood of SLE patients in association with marked
3.3.2. T cells in lupus nephritis upregulation of IL-17 and IL-23 concentrations, the latter being a
MRL/lpr mice with B cells which are capable of expressing surface cytokine involved in the maintenance of Th17 cells [187,188]. This
Ig, but not of secreting Ig, develop interstitial nephritis, rather than was also observed in children with LN. In addition, serum levels of
glomerulonephritis [169]. This suggests the existence of another am- IL-17 and IL-23 were found to correlate with the SLE disease activity
plification loop in which B lymphocytes act as antigen-presenting score, SLEDAI-2K [189]. Peripheral double negative (DN) T cells are
cells in order to activate T cells. As a matter of fact, the tubulo- also markedly expanded in subjects with SLE [189,190], and DN T
interstitial infiltrate in LN biopsy samples frequently contains T- and cells rather than TH17 (CD4+IL-17 +) T cells were found to be the
180 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

major source of IL-17 in these patients [190]. Importantly, both IL-17 reported between LN and the IRF7/KIAA1542 region (a polymorphism
and IL-23 were detected in 80% of the LN biopsy samples, mostly in in IRF7 is in strong linkage disequilibrium with an SNP in KIAA1542).
the tubulointerstitial region. In these biopsies, DN T cells were inter- Interestingly, these SNPs were not associated with SLE susceptibility
spersed with CD4 + and CD8 + T cells. This along with other experi- in this population [202], but were identified as a risk factor for SLE in
mental data implicates the role of IL-17 in the tissue injury of a European cohort [210].
glomerulonephritis [188,191].
4. Treatment
3.4. Genetics
4.1. Treatment of class I and II
The pathogenesis of SLE involves interactions between environ-
mental and genetic factors [192]. Deficiency of C1q is the strongest The type of therapy for LN depends on the severity of renal dis-
single genetic risk for developing SLE, but is responsible for only a ease. Patients with class I and II LN generally have a very good prog-
very small number of SLE cases. It is now recognized that multiple nosis and rarely require aggressive treatment for their renal disease,
genes, each making only a small contribution, account for the risk of and treatment is guided by other extra-renal SLE manifestations.
developing SLE. Several genome-wide scans have allowed major ad- However, optimal blood pressure control with angiotensin converting
vances in the identification of genetic regions predisposing to SLE, enzyme (ACE) inhibitors or angiotensin receptor blockers (ARB) is
bringing the total number of validated loci to more than 30 [193]. vital. These agents have the added advantage of reducing proteinuria,
Similar data are not yet available for LN, and most of the association and retrospective data suggest that they protect from the develop-
studies to date have yielded inconsistent results across populations. ment of biopsy proven glomerulonephritis [211,212]. Therefore, all
Susceptibility to SLE is consistently associated with HLA-DRB1*1501 LN patients with proteinuria ≥ 0.5 g/24 h should receive blockade of
and HLA-DRB1*0301, particularly in Caucasian populations [193]. An the rennin–angiotensin system [213].
association of LN with HLA-DRB1*15 has been noted in some studies
[30,193], and an interaction between DRB1*15 and DQA1*01 in 4.2. Treatment of class III and IV
enhancing susceptibility to LN has been suggested, but not yet
confirmed independently [194]. Interestingly, the DRB1*0301 allele Proliferative glomerulonephritis (classes III and IV) represents the
was protective against LN in univariate, but not in multivariate, anal- most severe manifestations of renal pathology and requires aggres-
ysis in a multiethnic cohort [30]. Among the most consistently identi- sive immunosuppressive therapy in order to prevent progressive
fied associations with SLE are the FCγRI, II and III genes. However, a loss of renal function and minimize associated morbidity and mortal-
recent meta-analyses indicate that the only significant association of ity. Corticosteroids have been the mainstay of every treatment regi-
LN with any FCGR polymorphism is seen with the F158 allele of men for proliferative LN since it was discovered in the 1960s that
FCγRIIIA-V/F158 in Asians only, but not in patients of European high doses (>40 mg prednisone) for prolonged periods of time
or African descent [195]. In contrast, the FCγRIIα-R/H131 and (>6 months) can prevent, or at least delay progression to renal fail-
FCγRIIIβ-NA1/NA2 polymorphisms demonstrate no association with ure [214]. Studies conducted in the 1970s and 1980s by the National
LN [196,197], whereas data on FCγRIIβ-T/I232 are limited [197]. Institutes of Health (NIH) established that the combination of cortico-
A variety of candidate genes in the type I IFN pathway have also steroids with cyclophosphamide (CYC) was superior to therapy with
been investigated for a possible association with LN, among them, corticosteroids alone for preserving renal function loss [215–217]. It
STAT4 that encodes a transcription factor which is activated by vari- was also noted that adding quarterly intravenous (iv) pulse CYC
ous growth factors and cytokines, including IFNα. STAT4 has been (ivCYC) to the maintenance regimen of oral corticosteroids for an ad-
identified as a risk factor for SLE in genome-wide scans in various ditional two years reduced the rate of LN exacerbations [218]. Similar
populations [193]. An association between a STAT4 haplotype and to cancer treatment, it is now a custom to distinguish between induc-
LN was seen in two large studies of patients of European ancestry tion and maintenance therapy. Induction therapy can be defined as
[198,199], though not in a smaller European study [200]. It also did “a period of intensive therapy with the aim of achieving a clinically
not quite reach statistical significance in a cohort of 308 Japanese meaningful and sustained response in a patient with active disease”
SLE patients [201], and was not detectable in a Han Chinese popula- [219]. The duration of induction therapy is generally individualized
tion [202]. This suggests that there may be ethnic differences in the and should be closely monitored for clinically meaningful response.
association between STAT4 genotype and SLE phenotype. The risk This depends on disease severity but should last at least 3 months
variant was found to correlate with higher transcript levels of STAT4 and should be extended to at least 6 months in cases of persistent
[203] and to enhance sensitivity to IFNα [140], providing biological disease activity. The definitions of “clinically meaningful response”
plausibility to the genetic association. Note, however, that STAT4 have been highly variable in published randomized controlled trials
deficiency in experimental animals can reduce or enhance the devel- (RCTs), making direct comparisons virtually impossible. Hopefully,
opment of glomerulonephritis depending on the precise genetic future clinical trials adhere to the guidelines proposed by the American
background, although autoantibody levels were significantly reduced College of Rheumatology (ACR) and the European League Against
in all models [204–206]. A recent genome-wide screen in Han Rheumatism (EULAR) [219,220] (summarized in Tables 2 and 3).
Chinese identified several novel genetic associations with SLE not Maintenance therapy can be defined as “a period of less intensive
previously identified in European populations [207], of which IKZF1 therapy following a period of induction therapy that has achieved a
was found to be particularly associated with LN [208]. This gene partial or complete response, with the aim of keeping the patient
encodes the Ikaros family zinc finger 1 transcription factor, which free of disease activity.” Note, however, that median time to remis-
has a role in lymphocyte differentiation and proliferation, in part by sion is approximately 10 months [221–223], meaning it is not
targeting STAT4 in T cells. uncommon for meaningful clinical responses to occur only during
Interferon regulatory factors (IRFs) are critical regulators of type I maintenance therapy. Therefore, the distinction between induction
IFN expression in response to TLR engagement, and the subsequent and maintenance therapy is somewhat arbitrary except for the
induction of many type I IFN-regulated genes. In addition, it is in- intensity of the therapeutic regimen.
creasingly recognized that IRFs have a wide array of other immune
functions. While IRF5 is clearly a risk factor for SLE [207,209], signifi- 4.2.1. The NIH and Euro-Lupus (ELNT) regimens
cant associations with LN have so far not been detected [199,200]. The standard regimen developed subsequent to the NIH studies
However, in a Han Chinese population, a strong association was consists of 6 monthly pulses of ivCYC at a target dose of 0.5–1 g/m 2
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 181

Table 2
Definitions of partial and complete responses formulated by the American College of Rheumatology (ACR) [220], European League Against Rheumatism (EULAR) [219], and the
Childhood Arthritis and Rheumatology Research Alliance (CARRA) [264].

ACR EULAR CARRA in juvenile LN

Mild response 30–50% improvement in 2 core renal


parameters (proteinuria, renal function as
creatinine clearance or serum creatinine, or
urine sediment) without clinically relevant
worsening of the remaining renal core
parameter
Improved/partial response/ 25% increase in estimated abnormal Proteinuria ≤0.5 g/daya At least 50% improvement in 2 core renal
moderate response baseline GFR Normal GFR (>90 ml/min) or, if parameters (with maximum spot protein/
≥50% reduction in the urinary protein: previously abnormal, improved to a stable creatinine ratio ≤1.0) without clinically
creatinine ratio to a value of 0.2–2.0 level within 10% of normal GFR relevant worsening of the remaining renal
Change from active urinary sediment (≥81 ml/min); core parameter
(>5 RBC/hpf and >5 WBC/hpf and/or ≥1 Inactive urinary sediment (≤5 RBC or
cellular casts) to inactive sediment WBC/hpf [or b10/mm3 or b10/μl]) and no
(≤5 RBC/hpf and ≤5 WBC/hpf and no cellular casts
cellular casts)
Complete response Estimated GFR of ≥90 ml/min/1.73 m2 Proteinuria ≤0.2 g/daya Normalization of renal function
and ≥50% reduction in the urinary protein: Normal GFR (>90 ml/min) or, if previously Inactive urine sediment (≤5 WBC/hpf,
creatinine ratio to a value of b0.2 and abnormal, improved to a stable level within b5 RBC/hpf, and no casts)
inactive urinary sediment 10% of normal GFR (≥81 ml/min); Spot protein/creatinine ratio b 0.2 or age
Inactive urinary sediment (≤5 RBC or WBC/ appropriate
hpf [or b10/mm3 or b10/μl] and no
cellular casts)
Remission A sustained response of at least 3–6 months,
but it cannot be judged to be a complete
remission in the absence of a biopsy

GFR = glomerular filtration rate.


hpf = high power field.
RBC = red blood cell.
WBC = white blood cell.
a
The expert group did not reach an absolute consensus on the values for proteinuria that constituted partial or complete responses, but the values given here reflect the majority
opinion.

body surface followed by pulses at 3-month intervals for 2 years. This is substantially higher in patients with SLE compared to the general
in combination with 3 initial pulses of iv methylprednisolone, followed population. The use of gonadotropin releasing hormone analogs dur-
by oral prednisone tapered from an initial dose of 0.5–1 mg/kg to a ing ivCYC therapy has been associated with preservation of ovarian
maintenance level of 5–10 mg/day [218] (see Table 4). While this function in the majority of treated patients [230,231]. These findings
regimen induces remission and preserves renal function in the ma- await confirmation in larger prospective studies, and the adverse
jority of the patients, it is also associated with severe and high event profile of such therapy needs to be evaluated. Male SLE fertility
rates of adverse events. Specifically, up to 25% of patients experience data is limited, but a small study did find that those men treated with
major infections [224,225], and herpes zoster occurs in up to 32% of ivCYC had more oligozoospermia, azoospermia and teratozoospermia
patients [215,216]. Hemorrhagic cystitis has also been reported than their counterparts [232].
with ivCYC, although much more common in patients on oral CYC Malignancies, severe leucopenias, and alopecia are other major
(as many as 17%) [226]. Many physicians routinely add Mesna to concerns with ivCYC therapy. To date, there is no clear evidence
the ivCYC regimen. Up to 60% of women with LN may experience that the efficacy of daily oral CYC differs significantly from that of
ovarian failure as a consequence of CYC treatment [215,217]. There pulse ivCYC [215,233]. However, because the studies at the NIH dem-
are indications that oral CYC carries a greater risk than ivCYC, possi- onstrated an increased risk of hemorrhagic cystitis and premature
bly due to the higher cumulative dose with oral therapy [215,226]. ovarian failure with oral CYC therapy [215], the iv route of adminis-
Both cumulative dose of CYC and age are strong determinants of tration is preferred, at least by physicians in North America and
sustained amenorrhea, with women past the age of 31 years being Europe.
at greatly increased risk [227–229]. Premature ovarian failure is not The side effect profile of pulse ivCYC and high dose corticosteroid
only devastating in a patient group that is dominated by young therapy led to the search of regimens with similar efficacy, but less
women in their childbearing years, but also increases the risk of toxic. Two approaches have been investigated: lowering the dose of
osteoporosis and cardiovascular disease, a risk that is already ivCYC and replacing ivCYC with other immunosuppressants either

Table 3
Definitions of flares formulated by the EULAR [219], and the Childhood Arthritis and Rheumatology Research Alliance (CARRA) [264].

EULAR [219] CARRA in juvenile LN [264]

Flare An increase in disease activity requiring a change of or


increase in therapy
Proteinuric flare Persistent proteinuria > 0.5–1.0 g/day after a complete response A persistent increase in proteinuria to values > 0.5 after
or a doubling of proteinuria, with values higher than 1.0 g/day achieving a complete response or a doubling of proteinuria
after a partial response with values > 1.0 after achieving a partial response
Nephritic flare Increase or recurrence of active urinary sediment (increased Increase or recurrence of active urinary sediment (increased
hematuria with or without reappearance of cellular casts) hematuria with or without reappearance of cellular casts)
with or without a concomitant increase in proteinuria with or without a concomitant increase in proteinuria
Severe nephritic flare Recurrence of active urinary sediment with an increase ≥25%
in serum creatinine
182
Table 4
Therapeutic regimens in RCTs comparing MMF and/or AZA for induction and/or maintenance to the NIH regimen. Note that all induction regimens are accompanied by oral corticosteroids, which are tapered to a maintenance level of
5–10 mg/day of prednisone or equivalent. The test drug is highlighted by bold print.

Cohort 3 iv MP pulses Induction Maintenance

n WHO class Drug Dose Schedule Duration Drug Dose Remarks Reference

NIH regimen 1 g/m2 ivCYC 0.5–1 g/m2 1 pulse per 6 months ivCYC Quarterly pulses [218]
body surface month for 2 more years
Euro-Lupus (Europe) 46 III, 750 mg ivCYC Max 1.5 g 1 pulse per 6 months ivCYC, then AZA 2 quarterly pulses Initial dose of ivCYC was 0.5 g, [225]
IV, month 2 mg/kg/day increased in 250 mg increments
Vc or AZA was started 2 weeks after the
Vd last ivCYC pulse
44 Low-dose ivCYC 0.5 g/m2 6 biweekly 10 weeks AZA AZA was started 2 weeks after the
pulses last ivCYC pulse
Miami, FL 20 III, 0 ivCYC NIH Up to 7 months ivCYC NIH Patients were switched to [224]
IV or maintenance therapy regardless of
Vb the response to the induction
regimen
20 NIH MMF 0.5–3 g/day
19 NIH AZA 1–3 mg/kg bw/

A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194


day
MAINTAIN (Europe) 53 III, 750 mg Low-dose ivCYC ELNT 6 biweekly 10 weeks MMF Target 2 g/day Randomization regardless of [258]
IV, (0.5 g/m2) pulses response at 3 months
52 Vc, AZA Target 2 mg/
Vd kg bw/day
a
ALMS (International) 116 III, ivCYC or MMF 24 weeks MMF 2 g/day Randomization only after achieving [259]
IV, or a pre-defined response to induction
V with either ivCYC or MMF
111 AZA 2 mg/kg bw/day
b
ALMS (International) 185 III, ivCYC NIH 1 pulse per 24 weeks [241]
IV, or month
185 V MMF Target 3 g/day 1.5 g, 24 weeks MMF was titrated from 1 g daily in
2× per day week 1, to 2 g daily in week 2, and to
3 g daily in week 3
US multicenter 69 III 0 ivCYC 24 weeks [242]
71 IV, or MMF Maximum 1 g, 24 weeks MMF was titrated from 1 g daily in
V 3 g/day 3× per day week 1, to 1.5 g daily in week 2, to a
maximum of 3 g daily thereafter
Malaysia multicenter 25 III, or 0 ivCYC 0.75–1 g/m2 1 pulse per 6 months [240]
IV month
19 MMF 2 g/day 1 g, 2× per day 6 months
Nanjing, China 23 IV 0.5–1 g ivCYC 0.75–1 g/m2 1 pulse per 6 months [239]
month
23 0 MMF 1–1.5 g/day 0.5–0.75 g, The dose of MMF was reduced to
2× per day 0.5–1 g/day after 3–6 months
depending on disease activity
DLNS (the Netherlands) 50 III 0 ivCYC 0.75 g/m2 1 pulse per 6 months ivCYC; followed by AZA 7 quarterly [252]
IV month pulses; 2 mg/kg/
Vc or day
37 Vd 1g AZA+MP 2 mg/kg/ AZA daily; AZA 2 mg/kg/day
day+ 1 g/day additional 3
iv MP daily Methyl
prednisone
pulses at weeks
2 and 6
Hong Kong 31 IV 0 Oral CYC 2.5 mg/kg/day daily 6 months AZA 1.5–2 mg/kg/day [247]
33 MMF 2 g/day 1 g, 2× per day MMF or AZA See remarks MMF dose was originally halved
after 6 months and replaced by AZA
after 12 months; later, patients re-
ceived 1.5 g/day for 6 months and
1 g/day for an additional 12 months
before being switched to AZA
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 183

during maintenance alone or during induction and maintenance — effective compared to MMF in patients whose ethnicity was classified
always in conjunction with glucocorticoids. The Euro-Lupus Nephritis as “other” (mostly of African, Mexican-mestizo and mixed ancestry)
Trial investigated whether the dose of ivCYC used as induction thera- [241]. Post hoc analyses revealed that Hispanics also had a higher
py could be lowered without losing efficacy [225] (see also Table 4). response rate to MMF compared to ivCYC, and that more patients of
Of note, another major departure from the NIH regimen in this trial African descent receiving ivCYC withdrew because of adverse events
was the use of azathioprine (AZA) for maintenance in both treatment compared to those receiving MMF. Opposite results were observed
arms. However, at the time the trial was designed (recruitment began for patients of European and Asian ancestry [241,245]. Interestingly,
in 1996), there had been no published randomized controlled trials Asians tolerated MMF more poorly and withdrew more frequently
establishing the equivalence of AZA and CYC in maintenance therapy, due to its adverse events compared to other racial groups. Thai pa-
even though AZA had been in clinical use for some time [234]. After a tients with LN were found to have low oral clearance of the active
median of 41 months of follow-up, there were no significant differ- metabolite of MMF [246], suggesting a possible explanation for this
ences between the two treatment arms in terms of the rates of treat- excess toxicity. Of note, the target dose of MMF was 3 g/day (in divided
ment failure, remissions and flares. Infections were twice as frequent doses) in this international RCT and the median dose was 2.6 g/day
in the high dose compared to the low dose group, but this did not [241], whereas earlier studies in Asian populations used as little as
reach statistical significance [225]. Similar results were reported 1–1.5 g/day in China and 2 g/day in Hong Kong and Malaysia with
after the 73 months of follow-up, with renal functional impairment remarkable efficacy and few discontinuations because of adverse events
being observed at similar rates in the two groups [235]. At ten [239,240,247]. Even doses of 0.5–1 g/day were reported to induce clin-
years, still no significant difference between the two treatment ically meaningful responses in Taiwanese patients with class III-V LN
regimens could be observed, with regards to death, end-stage renal [248]. Recent data from China indicate that MMF at a dose of 0.75–1 g
disease (ESRD), and sustained doubling of serum creatinine occurring twice daily is significantly more effective and less toxic than ivCYC
at similar rates [236]. When channeling these studies to clinical prac- even in the most severe forms of LN, namely class IV LN with
tice, one needs to take into consideration that the vast majority of non-inflammatory necrotizing vasculopathy [249], and in crescentic
the patients enrolled were of European ancestry; therefore, it may LN (ISN/RPS class IV or class IV+ V with crescent formation affecting
not be appropriate to extrapolate these findings to other patient >50% of glomeruli) [250]. Interestingly, although the actual drug cost
populations. Early data from a small prospective controlled study of MMF is considerably higher compared to ivCYC, the results of a
suggest that the NIH and the ELNT regimens are equally effective in patient-level simulation model suggest that induction therapy with
Egyptian patients [237]. MMF is actually less expensive compared to ivCYC. This is probably
due to the additional costs for the procedures and facilities that are nec-
4.2.2. Mycophenolate mofetil (MMF) as induction therapy essary for iv drug administration and the additional cost of antiemetics
The results of several small Asian studies demonstrate that the and Mesna as well as the higher rate of major infections with ivCYC
efficacy of MMF as induction treatment is similar to that of ivCYC compared to MMF [251].
[238–240]. However, a large international multi-center trial designed
to demonstrate the superiority of MMF failed to do so [241]. In con-
trast, a US multicenter non-inferiority trial actually demonstrated 4.2.3. AZA plus methylprednisolone for induction
the superiority of MMF compared to ivCYC as an induction therapy A Dutch RCT compared an induction treatment with 6 pulses of
since the rates of complete remission were significantly higher in ivCYC to a combination of AZA at 2 mg/kg/day plus iv pulses of 1 g/d
patients receiving MMF [242]. The frequency of infections in general of methylprednisolone for 3 days at week 0, 2 and 6, both regimens
and severe infections in particular was similar in two studies being combined with oral prednisone [252] (see also Table 4). Both
[240,241], but higher in the ivCYC-arm induction compared to MMF groups received AZA plus prednisone after 2 years. During the first
in another RCT [242]. Leucopenia appears to be somewhat more fre- two years of treatment, there was no significant difference in the cumu-
quent with ivCYC than with MMF [239,242]. Divergent results have lative incidence of partial or complete renal remission. However, repeat
been reported on gastrointestinal side effects, with some studies biopsies obtained after 2 years from a subset of participants in this trial
reporting a similar incidence of nausea and vomiting during ivCYC demonstrated that the chronicity score increased significantly more in
and MMF treatment [240,242], whereas others found these distur- the AZA group compared to ivCYC; the activity index decreased to the
bances to be significantly more frequent in patients treated with same extent in both groups [253]. After extended follow-up (up to a
MMF [241]. Diarrhea was markedly more frequent during induction median of 9.6 years), relapses were significantly more frequent in the
therapy with MMF [241,242]. AZA group, and sustained doubling of serum creatinine demonstrated
Some of these discrepancies in the results for both the efficacy and a similar trend [252,254]. There were no significant differences in mor-
toxicity of MMF compared to ivCYC may stem from the different eth- tality or development of ESRD. During the first 6 months of treatment,
nic distributions of the study participants. The US multicenter study infections (particularly herpes zoster) were more frequent in the AZA
included a high proportion of African-American as well as Hispanic arm. This may have been due to the higher steroid dose in this group.
population, but fewer Asian patients [242], whereas the international In a prospective assessment of participants in this RCT, health-related
study included ~ 40% European, 33% Asian, and 27% “other” patients quality of life (HRQoL) improved in both groups, particularly during
[241]. There is growing evidence that African American and Hispanic the first year, with only minor differences between the two treatment
patients have poorer responses to ivCYC compared to patients of arms [255]. The mean treatment burden, assessed on a 5-point Likert
European ancestry [243,244]. The most striking differences came scale, was significantly higher in the CYC group at 24 months. Unfortu-
from the international trial itself, where ivCYC was significantly less nately, studies comparing HRQoL during other treatment regimens

Notes to Table 4
ALMS = Aspreva Lupus Management Study; DLNS = Dutch Lupus Nephritis Study; NIH = National Institutes of Health; MP = methylprednisolone.
AZA = azathioprine.
CYC = cyclophosphamide.IV = intravenous.
MMF = mycophenolate mofetil.
RTC = Randomized controlled trials.
a
See the study by Appel et al. [241] below.
b
This study purports to have followed the NIH protocol as described by Boumpas et al. [218], but does not mention the 3 iv methylprednisolone pulses that are part of this
protocol.
184 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

have been based on recall several years after treatment, making their existing clinical trials was highly variable and not always specified.
results rather questionable [256,257]. Yet, it seems quite likely that patients who did not respond to other
therapeutic modalities or who initially responded but relapsed differ
4.2.4. MMF or AZA for maintenance from patients who are treatment-naïve. 4) The existing meta-
A single-center study compared quarterly pulses of ivCYC, MMF analyses do not take the ethnic composition of the patient groups
and AZA for maintenance therapy, patients receiving MMF had a mar- into account, a factor that has been shown to influence treatment
ginally higher probability of survival (p = 0.11), a lower rate of the responses [241,243–245].
composite endpoint of death or ESRD (p = 0.05) and were significant- The ACR recently published new guidelines for the treatment of
ly more likely to remain free of relapses compared to patients treated LN [213].
with quarterly pulses of ivCYC [224]. The results were similar for the For class III and IV LN, the Task Force Panel recommends an initial
AZA group, except that the difference in overall survival and three daily pulses of iv glucocorticoids (500–1000 mg). It considers
event-free survival (i.e., free of death or ESRD) compared to ivCYC ivCYC and MMF as equivalent for induction therapy, but recommends
achieved statistical significance (p = .02 and .009, respectively), the use of MMF as the first line agent in African American and Hispanic
while relapse-free survival was only marginally better (p = 0.12). patients. For Asians, the total daily dose of MMF should not exceed 2 g,
The rate of hospitalizations, the frequency of infections overall and whereas a target dose of 3 g/day is recommended for non-Asians. The
of major infections, and the incidence of sustained amenorrhea use of ivCYC following the ELNT protocol should be reserved for patients
were all significantly higher in the ivCYC compared to the other of predominantly European descent. People not responding to induc-
groups, and nausea and vomiting were also more frequent. tion with ivCYC or MMF after 6 months should be switched to
Direct comparisons between AZA and MMF as maintenance thera- the other agent. While the Task Force Panel does not recommend AZA
py have yielded somewhat inconsistent results. In a predominantly for induction, it considers AZA and MMF essentially equivalent for
European patient group, the two treatment modalities resulted in maintenance therapy; ivCYC is no longer considered an option for
very similar outcomes (time to renal flare, severe systemic flare, or maintenance. The ACR guidelines specifically point out that there
renal remission) [258]. The frequency of adverse events in general currently is inadequate data to guide physicians as to when and how
as well as individual adverse events did not differ significantly be- to taper or withdraw AZA or MMF.
tween the two groups except for leucopenias and other cytopenias, Importantly, while MMF and CYC are teratogenic, AZA is associated
which were more frequent during AZA treatment. In contrast, others with a relatively low risk of fetal abnormalities and can be used as a
found MMF to be superior to AZA in terms of time to treatment failure steroid-sparing agent for active LN during pregnancy. Childhood-onset
overall and individual components of this outcome measure, i.e., time LN is not given special consideration in the ACR guidelines, even though
to renal flare and time to rescue therapy [259]. Serious adverse events the benefits of immunosuppressive therapies need to be weighed even
tended to be more common in the AZA group, and the frequency more carefully against their toxicities in the context of the increased
of withdrawals due to adverse events was significantly higher in vulnerability of a growing organism. Nonetheless, a recent survey
the AZA group. The trials differed in their induction regimens revealed that members of the Childhood Arthritis and Rheumatology
(Euro-Lupus vs. NIH or MMF) and in the requirement for a response Research Alliance most commonly used the NIH regimen of ivCYC for
to induction before randomization (see Table 4 for details). In addition, induction in patients with juvenile-onset LN [264]. It would seem advis-
the second study included 33.5% Asian patients, 43.6% of European able, however, to consider the Euro-Lupus regimen in order to minimize
ancestry, 10% black and 13% other [259]. It is difficult to determine drug-induced morbidity. A modified version of this regimen (6 monthly
to what extent these differences contributed to the outcome disparities. rather than biweekly pulses of 500 mg/m2 of ivCYC) followed by AZA,
It is customary to adjust the dose of ivCYC according to the white or MMF if not responsive to AZA. Good results have been reported
blood cell nadir 7–10 days after an infusion, with the minimum value with this regimen in Turkish children with class III or IV LN, although
requiring a dose reduction of 25% varying between 1500 and a comparison group is unfortunately missing [265]. Data are beginning
3500 cells/μl in different studies [218,224,240,252]. In contrast, to emerge demonstrating that tacrolimus may be as effective as ivCYC
there is no suggested dose adjustment regimen with MMF. Several for induction therapy in proliferative LN [266], while cyclosporine A
studies in patients with SLE, including mostly subjects with LN, have demonstrated similar efficacy as AZA in reducing proteinuria and pre-
shown that there is up to 10-fold inter-subject variability in indi- serving renal function during maintenance therapy [267]. The adverse
vidual plasma concentrations of mycophenolic acid (MPA), the active event profile was more favorable with tacrolimus and cyclosporine A
metabolite of MMF [246,260–263]. In analogy to the use of pharmaco- compared to ivCYC and AZA, respectively. Further studies are in prog-
kinetic parameters for individualizing doses of MPA in transplant ress with these agents as single therapy or in combination with MMF.
recipients, a Bayesian estimator of the area under the curve from However, these agents are not included in the ACR Task Force recom-
0 to 12 h (AUC0-12) of plasma MPA concentration has been derived mendations, likely because of insufficient evidence at this time.
for determining MPA exposure in SLE patients [260]. The resulting Several studies found that hydroxychloroquine decreases the risk of
AUC0-12 estimate was found to correlate inversely with disease progression to ESRD and mortality rate [268–270]. This is consistent
activity as assessed by SLEDAI or BILAG [261]. Based on this, an with the ability of antimalarials to enhance the response to immuno-
AUC0-12 of ≥ 35 mg h/l was identified as the target value above suppressive therapy with MMF, reduce the frequency of flares and
which active SLE becomes rather unlikely (negative predictive value limit damage accrual, including renal damage [271–274]. Consequently,
of 92%). In a pilot study in patients with childhood-onset LN, a similar the ACR treatment guidelines recommend adding hydroxychloroquine
value was identified (>30 mg h/l) [263]. Most available data is on to the regimen of all SLE patients with LN [213].
French and US patients, including a considerable portion of patients
with African ancestry, but few Asian subjects. 4.3. Treatment of membranous LN
Various “meta-analyses” on the efficacy of MMF compared to CYC
or AZA have been published, but have serious limitations. 1) They do Class V or membranous LN (MLN) often occurs in combination with
not take into account that almost every one of the clinical trials with proliferative lesions in which case it requires the same treatment as for
MMF used different definitions for outcome measures (e.g., partial or class III or IV lesions. Therapy of pure MLN remains somewhat contro-
complete remission, improvement, relapse). 2) The duration of versial, because renal functional decline is slower compared to class III
follow-up was highly variable [215,252]. The ACR now recommends or IV LN and the long-term prognosis is felt to be more favorable. That
a minimum trial duration “of 6 months, better 1 year, and ideally being said, MLN is often accompanied by nephrotic-range proteinuria,
5 years” [220]. 3) The proportion of newly diagnosed patients in which increases the risk of thromboembolic and cardiovascular
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 185

complications. Corticosteroids represent the mainstay of therapy in with MMF or ivCYC [213]. A phase III clinical trial investigating the
membranous LN, even though their benefit has not been clearly treatment of LN with the fully humanized anti-CD20 monoclonal
established. It is generally agreed that patients with high-grade protein- antibody ocrelizumab in addition to corticosteroids and MMF or
uria require the addition of other immunosuppresants to corticoste- ivCYC is ongoing (www.clinicaltrials.gov).
roids. Unfortunately, RCTs in patients with pure MLN are rare. There
are indications that induction and maintenance with ivCYC, MMF, and 4.4.3. Abatacept and other biologicals
AZA are effective in MLN [275,276], but cyclosporine A, tacrolimus, or Abatacept is a soluble Fc:CTLA-4 fusion protein that prevents T cell
chlorambucil may be similarly beneficial and less toxic [277–279]. activation by competing with CD28 for binding to CD80/86, a
Therefore the potential risks and benefits of each of these treat- costimulatory signal required for full T cell activation. In the first
ments must be carefully weighed in each individual patient. The ACR double-blind RCT of abatacept, none of the primary or secondary end-
recommends a combination of prednisone (0.5 mg/kg/day) and MMF points were met [287]. Patients with LN were specifically excluded
(2–3 g/day) for induction in patients with pure class V nephritis and from this trial. A phase II/III study of abatacept (plus MMF and corti-
nephrotic-range proteinuria [213]. costeroids) was terminated due to inefficacy in the short-term
phase (www.clinicaltrials.gov). Another phase two trial is ongoing.
4.4. Biological agents as further treatment options Overall the data are disappointing [288]. Trials with other biological
agents, such as epratuzumab, atacicept, and abetimus have either ex-
4.4.1. Belimumab cluded patients with LN, failed to demonstrate effectiveness, or are
Belimumab represents the first new drug approved for treatment of still ongoing. TNF antagonists or IL-1 blockade is not advisable in pa-
SLE in decades both by the Food and Drug Administration (FDA) and by tients with LN [285].
the European Medicines Agency. It is a fully human monoclonal anti-
body against B lymphocyte stimulator (BLyS), also known as B cell acti- 5. Biomarkers
vating factor (BAFF), a cytokine that provides survival signals for B cells
and is overexpressed in patients with SLE. Inhibition of BLyS with It would be of great help to use biomarkers that could obviate the
belimumab results in reductions in subsets of circulating CD20+ B need for biopsies, in particular repeat biopsies, in order to assess LN
lymphocytes and short-lived plasma cells. Two phase III trials of activity or kidney damage, and that could accurately predict an immi-
belimumab in addition to stable treatment with prednisone, anti- nent exacerbation of renal disease [289,290]. The usual marker of
malarials, or other immunosuppressants (but not ivCYC) in the treat- renal disease is urinary protein, but the currently recommended
ment of patients with active SLE demonstrated it to be effective in spot urinary protein to creatinine ratio may not be sufficiently sensi-
reducing disease activity and severe flares [280,281]. Patients with tive to detect early nephritis [220,291]. Conventional markers of ac-
severe active LN were specifically excluded from both trials, but signif- tive renal disease are serum levels of anti-dsDNA, anti-C1q antibody
icant effects on normalization of anti-dsDNA antibodies and low com- and complement levels. However, these have widely variable sensi-
plement levels suggest that belimumab may also be beneficial in LN. tivity depending on the assay system used, and their specificity
Post-hoc analysis of the few patients with proteinuria > 2 g/24 h indi- ranges between 50 and 75% for active renal disease [291,292]. Al-
cated that the proportion dropped from 9% and 7% to 5.5% and 4.2% in though both serum and urinary compounds have been investigated
the groups treated with 1 and 10 mg/kg belimumab, respectively. for their potential use as biomarkers, it is generally felt that urinary
Comparison with the placebo group (7.6% proteinuria> 2 g/24 h) did substances are likely to reflect kidney damage better. Urinary candi-
not meet statistically significant differences. However, there was a dates have included adhesion molecules, cytokines, chemokines,
significant reduction in the frequency of renal flares and a tendency and their receptors, including VCAM-1, P-selectin, IL-6, IP-10,
for greater reduction in proteinuria. RANTES, MCP-1, CXCL16, CX3CL1, TWEAK, and TNFR1 [98,291]. In
addition, proteomic approaches have identified a variety of other uri-
4.4.2. Rituximab and ocrelizumab nary proteins that are associated with LN, including transferrin, ceru-
Rituximab is a chimeric monoclonal antibody to CD20, which loplasmin, hepcidin 20, hepcidin 25, α1-acid-glycoprotein, lipocalin-
depletes B lymphocyte precursors and mature B cells because they type prostaglandin D-synthetase, and neutrophil gelatinase associated
express CD20 on their cell surface. A phase III trial examined the lipocalin. In particular, the second group of proteins (non-immune
effects of treatment with rituximab on days 1, 15, 168, and 182 com- related) contains some of the most promising candidates for
pared to placebo, both in combination with MMF, in patients with predicting renal flares [293,294]. However, the current results await
newly diagnosed LN [282]. The response rate was small and did not independent confirmation in large scale prospective studies. In addi-
meet statistical significance. While rituximab may not be more effec- tion, it is unlikely that a single biomarker will have sufficient sensitiv-
tive than standard care with immunosuppressive agents as first line ity and specificity for the diagnosis, activity, response to treatment, or
therapy, open label evidence suggests that rituximab has a place prognosis of LN. Therefore, future research should focus on the utility
in the treatment of patients with refractory disease [283–285]. of different biomarker combinations to evaluate active renal disease
According to a pooled data analysis from 164 patients with LN, most or to provide prognostic information.
of whom refractory to treatment, 27% of these patients achieved a
complete response with rituximab [284]. It appeared particularly 6. Outcome/prognosis
effective in patients with mixed membranous-proliferative LN and
type III LN (with complete response rates of 75% and 48%, respective- 6.1. Remission and flares
ly, compared to 27% in type V and 16% in type IV LN). Keeping the
rituximab risk profile in mind, there have been at least 3 reported A major outcome measure in clinical trials and observational stud-
cases of progressive multifocal leukoencephalopathy (PML), a gener- ies has been the achievement of “remission.” It should be noted, how-
ally fatal demyelinating disease, in patients with rheumatoid arthritis, ever, that the terms “major clinical response” or “partial clinical
and 2 reported cases in SLE patients with post treatment. Patients response” are preferable since true remission can only be established
with SLE and possibly other connective tissue diseases, but not with by a follow-up biopsy, which is not routinely performed [295]. None-
rheumatoid arthritis, have an increased risk of PML even in the ab- theless, we use the terms interchangeably in this review. In clinical
sence of biological therapy [286]. The FDA has issued a public health trials, approximately 50% of patients with proliferative LN experience
advisory on this issue. The ACR treatment guidelines state that a clinically meaningful response within 6 months, although complete
rituximab can be used in patients refractory to induction therapy responses are quite rare during induction therapy [239–242]. The
186 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

proportion of patients who eventually reach partial or complete re- significant in univariate analysis only and to lose significance once
mission ranges from ~ 60 to 90% in clinical trials and observational poverty, insurance status, various clinical factors, and non-compliance
studies [224,225,247,252,259]. Similar response rates have been were isolated [28,313–318]. In some multivariate analysis, Hispanic
reported in patients with membranous LN [296,297], with the excep- ethnicity was identified as a risk factor for doubling serum creatinine
tion of a Spanish cohort, where only 32% of patients with class V LN and progression to ESRD [28,314]. In contrast, Hispanic children in
achieved remission compared to 78% and 70% of patients with class Florida were significantly less likely to progress to ESRD [312]. This
III and class IV lesions, respectively [268]. Much of the variability underscores again the heterogeneity of “Hispanic” populations [29].
between studies is attributable to the use of different definitions of Overall, it seems likely that both socioeconomic status and “ethnicity”
remission, but also to different patient characteristics and treatment represent genetic as well as socio-cultural factors and contribute to
regimens. Median time to remission in patients with proliferative the worse outcome of LN in African-American and other non-
GN treated with ivCYC is approximately 10 months [221–223], European populations. Of note, in a predominantly African-American
although markedly shorter response times have been reported [247]. and Hispanic cohort of pediatric patients with LN, the introduction of
Kidney disease relapse even in a patient who achieved a complete MMF into the standard therapeutic regimen was associated with signif-
clinical response is common. Unfortunately, relapse rates are often icant improvement in 5-year renal survival from 52% to 91% [312]. This
reported as the proportion of patients experiencing a renal flare, may be attributable to the superior efficacy of MMF in these populations
which makes comparison difficult due to varying lengths of follow- [241,245].
up. However, it seems that at least 40% of patients with proliferative A long list of other demographic, clinical, and histopathological risk
GN will eventually have a flair [222,223,298,299], and only slightly factors for poor renal outcomes has been identified. Given the heteroge-
lower rates have been reported in MLN [296,297,300]. Median times neity of the disease itself, the investigated populations, and the treat-
to renal flare range from 32 to 79 months [222,298,299]. The induc- ment regimens, it is not surprising that the results have been
tion of re-remission after a renal exacerbation is more difficult and inconsistent. As expected though, treatment resistance has been identi-
successful in only about two-thirds of the patients [222]. Those who fied as an independent predictor of ESRD in numerous cohorts
fail to achieve a second remission are at increased risk of progressing [55,223,226,309,316,319], including patients with MLN [296,300].
to renal insufficiency or ESRD [222,298,299]. Predictors of renal flares Demonstrating a response within 6 months of initiation of therapy
include failure to achieve a complete response [223,299], persistence may be a particularly important predictor for good long-term outcome
of low complement levels after treatment [298,299], longer time to [235,236]. A renal flare represents new tissue injury and is likely to
remission [222], longer delay before the initiation of treatment result in renal cell death and fibrosis, leading to loss of glomerular and
[223], and African ancestry [298]. tubular function. Therefore, an association between renal relapses and
progression to ESRD is to be expected and has been found in several
6.2. Progression to ESRD studies of patients with both proliferative and membranous nephritis
[300,306,309,320], although statistical significance is not always
Although more treatment options have become available for LN retained in multivariate analysis [296,299]. Hypertension is commonly
over the last decades, analysis of data from the US Renal Data System found to predict progression to ESRD [305,313,321–324], and decreased
revealed a steady increase in the rate of ESRD due to LN between renal function at baseline is another frequently identified risk factor for
1982 and 1995 [301] and a subsequent stabilization, but no decline, poor renal outcome [55,223,268,300,305,313,321–326]. Diagnostic
of this rate in the years 1996–2004 [302]. A study using the same delay is also an important predictor of ESRD [327–329]. This suggests
data source, but adjusting more thoroughly for annual population that responsiveness to therapy may be decreased once renal function
changes and also investigating changes by US region, found that the has begun to deteriorate, possibly due to delayed diagnosis of LN and
incidence of ESRD actually rose in patients aged 5–39 years old, in progressive tissue damage. This is supported by the finding that a
African Americans and Native Americans, and in the southern US lower chronicity score for renal biopsy samples predict better response
between 1995 and 2006 [303]. That being said, this trend may actual- to treatment (with ivCYC) [60,226]. In addition, a significant indepen-
ly reflect increased survival rates in SLE patients. It may also be due to dent association has been detected between the chronicity index of
changes in the SLE epidemiology in general and/or the proportion of tubulointerstitial disease and progression to ESRD [178,179,300,305,
SLE patients with LN, but unfortunately, recent population-based 306,313,321,330]. Interestingly, class IV LN, which is generally thought
incidence data are not available for the US. Importantly, survival dur- to carry the greatest risk of progression to ESRD, is not consistently
ing the first 3 years of ESRD did not improve between 1995 and 2006 identified as an independent predictor of renal failure [56,305,322–
[303]. Comparisons of patient groups diagnosed and treated at the 324,327,331].
same institutions during different periods also demonstrate that the
rate of ESRD has not changed significantly over the last 30 years 6.3. Renal replacement therapy treatment
[304–306]. The time from ESRD to death increased only slightly and
non-significantly [304]. However, in one of these cohorts, patients Once a patient with LN has progressed to ESRD, the initial choices
with proliferative LN diagnosed between 1987 and 2000 faced a for renal replacement therapy include preemptive kidney transplan-
significantly lower risk of doubling serum creatinine compared to tation, peritoneal dialysis, or hemodialysis. Preemptive kidney trans-
patients diagnosed between 1973 and 1986 [306]. Some recent plantation carries a better graft and patient survival in recipients with
renal survival rates are summarized in Table 5. LN compared to dialysis treatment prior to transplantation [332].
It is amply documented that SLE patients from most non-European Compared with hemodialysis replacement therapy, peritoneal dialy-
populations develop renal involvement more frequently than patients sis prior to transplantation is associated with better allograft survival,
of European descent, are more often treatment resistant (at least to particularly in LN patients [333]. However, pediatric SLE patients who
ivCYC) and face a considerably poorer prognosis, i.e., develop renal are treated with peritoneal dialysis prior to kidney transplant have a
insufficiency and ESRD far more frequently and are at increased risk of significantly higher rate of early graft failure than patients who
death [11,29,243,307–309]. There have been numerous attempts to were treated with hemodialysis [334].
determine whether this is due to socioeconomic, sociocultural or In the US, the initial choice of renal replacement therapy for
other factors, but the results have been controversial. Several analyses patients with ESRD due to LN is strongly influenced by socio-
demonstrate African ancestry to be an independent risk factor for demographic factors, insurance status, and employment status
poor renal outcome [243,310,311], and this includes patients with [335], although this does not appear to differ greatly from the experi-
juvenile-onset LN [312]. Other studies, however, found it to be ence of patients with ESRD due to other causes [336]. Patients of
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 187

Table 5
Patient and renal survival.

Study location Study period n Classes Patient survival Renal survival Reference

5-year 10-year 15-year 20-year 5-year 10-year 15-year 20-year

Adults Barcelona, Spain 1970–2006 190 II–VI 92 80 [268]


Hong Kong 1968–2008 230 I–V 98.6 98.2 90.5 99.5a 98 89.7 [344]
Hong Kong 1988–2002 268 IV 88.7 82.8 70.7 [226]
Hong Kong 1976–1997 183 I–V 98.9 94.4 94.4 92.1 81.2 75.2 [368]
Copenhagen, Denmark 1971–1995 100 II–V 87 83 73 [305]
Zhenzhou, China 2003–2010 491 I–VI 88 77 53 45 [269]
Lucknow, India 1988–2007 188 II–VI 91 81 76 [323]
Riyadh, Saudi Arabia 1980–2006 299 I–VI 96 95 [324]
Italy 1974–2008 67 V (pure) 97.5 94.5 96.5 85.8 83 [300]
Nanjing, China 1986–2004 100 V 98 98 96.1 92.7 [296]
Children Taiwan 1979–1991 167 II–V 91.1 93.1a [369]
Serbia and Montenegro 1983–2001 53 I–V 98.1 88.6 [346]
Egypt ??–2007 100 II–V 82.4 64.7 [18]
India 1985–2007 180 I–V 77 65 53 92a 91a 79a [331]
a
Renal survival was calculated only for patients alive at the time of analysis.

African, Asian or Hispanic descent were found to be less likely to and 2008 [344], and 6.8 for a Danish cohort with all classes of LN
receive preemptive kidney transplants [335]. In addition, African followed from 1971 to 1995 [305]. Importantly, both studies found
Americans, Medicaid recipients and unemployed patients received that the SMRs had not changed significantly at least since the begin-
peritoneal dialysis less frequently compared to Americans of European ning of the 1980s [305,344], confirming the findings of an earlier
descent, privately insured and employed patients. Patients receiving study [345], although others found that survival improved into the
hemodialysis for ESRD secondary to LN are at increased risk of death 1990s and then stabilized [304]. Five-year survival rates > 98% have
compared to patients with ESRD due to other causes [337]. In particular been reported in adult and pediatric cohorts of patients with LN
the mortality risk for pediatric patients was found to be 2.4-fold higher [344,346], although it is not always clear whether diagnosis of SLE
compared to other children with ESRD. The risk of death while on or LN represents the starting point of the analysis. However, survival
hemodialysis was found to be markedly higher for African American rates remain markedly lower in developing countries, particularly in
children with ESRD due to LN [338]. In addition, African American and pediatric cohorts (see also Table 5). The difference becomes more
Hispanic patients with childhood-onset LN and ESRD had 50% and 37% pronounced with longer follow-up, with recently reported 10-year
lower 5-year survival rates of kidney transplantation compared to survival rates ranging from 68% to 98.2% [9,344,347].
other populations even after adjusting for US region and medical insur- In a cohort from Hong Kong, the SMR for patients who developed
ance, which were also significantly associated with the rate of kidney ESRD was 26.1, even though ESRD was the immediate cause of death
transplantation. in only one of the 24 patients who died [344]. However, chronic as
Most large-scale studies demonstrate equivalent renal and patient well as acute renal failure remains an important cause of death for pa-
survival in patients with ESRD due to LN compared to patients with tients with LN in developing countries [9,324,331]. Generally infec-
ESRD due to other causes [339,340], and limited data suggest that tions represent the leading cause of death in adult and pediatric
the same is true for adult patients with childhood onset of LN [334]. patients with LN in industrialized and developing countries around
African-American transplant recipients, but not Hispanic recipients, the world [226,268,269,312,323,324,344]. This is most likely due to
were at significantly increased risk of graft failure, but not of death, the long-term use of immunosuppressive therapies in these patients.
in a multivariate analysis of data from the US Renal Data System be- Cardiovascular and cerebrovascular disease accounts for about
tween 1995 and 2002 [333]. Possible explanations for higher preva- 15–30% of all deaths in patients with LN, reflecting the increased
lence of allograft failure in African American transplant recipients risk of cardiovascular complications in patients with SLE overall, as
are higher rates of cadaveric allograft, a higher percentage of two well as the combined harmful effects of renal failure and chronic
HLA loci mismatches, experiencing more delayed graft function, and corticosteroid therapy [226,268,269,312,344]. Treatment with CYC is
more early rather than late rejection episodes [341,342]. In LN known to increase the risk of malignancies, and these are responsible
renal allograft recipients, recurrence of LN is seen in about 2–30% for approximately 10% of deaths in LN cohorts [226,268,344].
[341–343]. It is generally mild (most often mesangial, less frequently
membranous, and rarely proliferative), and was found to be a risk fac- 6.5. Future directions
tor for allograft loss, but does not significantly affect patient survival
[342]. The only significant risk factor for LN recurrence identified in It is clear that there remains a major void in the successful man-
a multivariate analysis was African-American ancestry [342]. agement of lupus nephritis. On the other hand, the strides in the
basic biology of lupus in both humans and mice have been striking.
6.4. Patient survival Examples of basic observations that need eventually to be translated
into clinical therapeutics include detailed understandings of regulato-
Starting in the 1950s, the survival of SLE patients overall increased ry T cells, the correlation, if any, between the multiple variants of mu-
from less than 50% to >95% at 5 years post diagnosis in industrialized rine lupus and human disease, the increasing interest in epigenetics,
areas [6]. Nonetheless, the standardized mortality ratio (SMR) gener- the role of environmental agents, including drugs, in disease induc-
ally remains ≥ 2, even if survival of lupus patients approaches that of tion, methodology to alter complement activation in kidneys, defini-
the general population in some regions [7]. Although the risk of death tion of the critical roles of toll-like receptors, a better understanding
from renal disease has decreased substantially over the last decades, of hematopoietic precursors and their role in loss of tolerance, more
mortality remains significantly higher among patients with LN com- aggressive use of genomics, study of critical populations, including
pared to patients with SLE overall [7]. The SMR (reflecting all causes children, identification of new biomarkers, contemporary translation
of death rather than renal disease alone) was 5.9 for patients with of the immunobiology of tolerance in mice to humans, a better under-
proliferative LN who were followed in Hong Kong between 1968 standing of trafficking, the role of apoptosis in augmenting the
188 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

immune response, the use of peptides and other therapeutic agents to [23] Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, et al. Systemic
lupus erythematosus: clinical and immunologic patterns of disease expression
alter immune cell interactions and the role of innate immunity and in a cohort of 1,000 patients. The European Working Party on Systemic Lupus
autoinflammation [4,5,34,40,177,348–367]. Erythematosus. Med (Baltimore) 1993;72:113-24.
[24] Brugos B, Kiss E, Szodoray P, Szegedi G, Zeher M. Retrospective analysis of
patients with lupus nephritis: data from a large clinical immunological center
Take-home messages in Hungary. Scand J Immunol 2006;64:433-7.
[25] Burgos PI, McGwin Jr G, Pons-Estel GJ, Reveille JD, Alarcon GS, Vila LM. US
• Lupus nephritis remains a major challenge and continues to be one patients of Hispanic and African ancestry develop lupus nephritis early in the
disease course: data from LUMINA, a multiethnic US cohort (LUMINA LXXIV).
of the more severe manifestations of SLE. Ann Rheum Dis 2011;70:393-4.
• Although substantial research has helped define the pathogenetic [26] Hopkinson ND, Jenkinson C, Muir KR, Doherty M, Powell RJ. Racial group, socio-
mechanisms of lupus nephritis, such research has not adequately economic status, and the development of persistent proteinuria in systemic
lupus erythematosus. Ann Rheum Dis 2000;59:116-9.
translated into new therapeutic agents.
[27] Bastian HM, Roseman JM, McGwin Jr G, Alarcon GS, Friedman AW, Fessler BJ,
• Early diagnosis and intervention are essential for favorable patient et al. Systemic lupus erythematosus in three ethnic groups. XII. Risk factors for
outcomes. lupus nephritis after diagnosis. Lupus 2002;11:152-60.
[28] Alarcón GS, McGwin Jr G, Petri M, Ramsey-Goldman R, Fessler BJ, Vilá LM, et al.
Time to renal disease and end-stage renal disease in PROFILE: a multiethnic
lupus cohort. PLoS Med 2006;3:e396.
References [29] Pons-Estel BA, Catoggio LJ, Cardiel MH, Soriano ER, Gentiletti S, Villa AR, et al.
The GLADEL multinational Latin American prospective inception cohort of
[1] Doria A, Zen M, Bettio S, Gatto M, Bassi N, Nalotto L, et al. Autoinflammation and 1,214 patients with systemic lupus erythematosus: ethnic and disease heteroge-
autoimmunity: bridging the divide. Autoimmun Rev 2012;12:22-30. neity among “Hispanics”. Med (Baltimore) 2004;83:1–17.
[2] Singh RP, Waldron RT, Hahn BH. Genes, tolerance and systemic autoimmunity. [30] Bastian HM, Alarcón GS, Roseman JM, McGwin Jr G, Vilá LM, Fessler BJ, et al. Sys-
Autoimmun Rev 2012;11:664-9. temic lupus erythematosus in a multiethnic US cohort (LUMINA) XL II: factors
[3] Hiraki LT, Feldman CH, Liu J, Alarcon GS, Fischer MA, Winkelmayer WC, et al. predictive of new or worsening proteinuria. Rheumatology (Oxford) 2007;46:
Prevalence, incidence, and demographics of systemic lupus erythematosus and 683-9.
lupus nephritis from 2000 to 2004 among children in the US Medicaid beneficia- [31] Deshmukh US, Sim DL, Dai C, Kannapell CJ, Gaskin F, Rajagopalan G, et al.
ry population. Arthritis Rheum 2012;64:2669-76. HLA-DR3 restricted T cell epitope mimicry in induction of autoimmune response
[4] Mok CC, Kwok CL, Ho LY, Chan PT, Yip SF. Life expectancy, standardized mortality to lupus-associated antigen SmD. J Autoimmun 2011;37:254-62.
ratios, and causes of death in six rheumatic diseases in Hong Kong, China. Arthritis [32] Jorgensen KT, Pedersen BV, Nielsen NM, Jacobsen S, Frisch M. Childbirths and
Rheum 2011;63:1182-9. risk of female predominant and other autoimmune diseases in a population-
[5] Watson L, Leone V, Pilkington C, Tullus K, Rangaraj S, McDonagh JE, et al. based Danish cohort. J Autoimmun 2012;38:J81-7.
Disease activity, severity, and damage in the UK juvenile-onset systemic lupus [33] Fu SM, Deshmukh US, Gaskin F. Pathogenesis of systemic lupus erythematosus
erythematosus cohort. Arthritis Rheum 2012;64:2356-65. revisited 2011: end organ resistance to damage, autoantibody initiation and
[6] Borchers AT, Keen CL, Shoenfeld Y, Gershwin ME. Surviving the butterfly and the diversification, and HLA-DR. J Autoimmun 2011;37:104-12.
wolf: mortality trends in systemic lupus erythematosus. Autoimmun Rev [34] Kontaki E, Boumpas DT. Innate immunity in systemic lupus erythematosus:
2004;3:423-53. sensing endogenous nucleic acids. J Autoimmun 2010;35:206-11.
[7] Bernatsky S, Boivin JF, Joseph L, Manzi S, Ginzler E, Gladman DD, et al. Mortality [35] Scheinecker C, Bonelli M, Smolen JS. Pathogenetic aspects of systemic lupus
in systemic lupus erythematosus. Arthritis Rheum 2006;54:2550-7. erythematosus with an emphasis on regulatory T cells. J Autoimmun 2010;35:
[8] Rabbani MA, Habib HB, Islam M, Ahmad B, Majid S, Saeed W, et al. Survival analysis 269-75.
and prognostic indicators of systemic lupus erythematosus in Pakistani patients. [36] Alarcón GS, Bastian HM, Beasley TM, Roseman JM, Tan FK, Fessler BJ, et al. Systemic
Lupus 2009;18:848-55. lupus erythematosus in a multi-ethnic cohort (LUMINA) XXXII: [corrected] contri-
[9] Flower C, Hennis AJ, Hambleton IR, Nicholson GD, Liang MH. Systemic lupus butions of admixture and socioeconomic status to renal involvement. Lupus
erythematosus in an Afro-Caribbean population: incidence, clinical manifesta- 2006;15:26-31.
tions and survival in the Barbados National Lupus Registry. Arthritis Care Res [37] Andrade RM, Alarcon GS, Fernandez M, Apte M, Vila LM, Reveille JD. Accelerated
2012;64:1151-8. damage accrual among men with systemic lupus erythematosus: XLIV. Results
[10] Pons-Estel GJ, Serrano R, Plasin MA, Espinosa G, Cervera R. Epidemiology and from a multiethnic US cohort. Arthritis Rheum 2007;56:622-30.
management of refractory lupus nephritis. Autoimmun Rev 2011;10:655-63. [38] Austin III HA, Muenz LR, Joyce KM, Antonovych TA, Kullick ME, Klippel JH, et al.
[11] Borchers AT, Naguwa SM, Shoenfeld Y, Gershwin ME. The geoepidemiology of Prognostic factors in lupus nephritis. Contribution of renal histologic data. Am
systemic lupus erythematosus. Autoimmun Rev 2010;9:A277-87. J Med 1983;75:382-91.
[12] Patel M, Clarke AM, Bruce IN, Symmons DP. The prevalence and incidence of [39] Molina JF, Drenkard C, Molina J, Cardiel MH, Uribe O, Anaya JM, et al. Systemic
biopsy-proven lupus nephritis in the UK: evidence of an ethnic gradient. Arthritis lupus erythematosus in males. A study of 107 Latin American patients. Medicine
Rheum 2006;54:2963-9. (Baltimore) 1996;75:124-30.
[13] Abdwani R, Rizvi SG, El-Nour I. Childhood systemic lupus erythematosus in Sultan- [40] Sawalha AH, Wang L, Nadig A, Somers EC, McCune WJ, Hughes T, et al. Sex-
ate of Oman: demographics and clinical analysis. Lupus 2008;17:683-6. specific differences in the relationship between genetic susceptibility, T cell
[14] Barron KS, Silverman ED, Gonzales J, Reveille JD. Clinical, serologic, and immuno- DNA demethylation and lupus flare severity. J Autoimmun 2012;38:J216-22.
genetic studies in childhood-onset systemic lupus erythematosus. Arthritis [41] Voulgari PV, Katsimbri P, Alamanos Y, Drosos AA. Gender and age differences in
Rheum 1993;36:348-54. systemic lupus erythematosus. A study of 489 Greek patients with a review of
[15] Font J, Cervera R, Espinosa G, Pallares L, Ramos-Casals M, Jimenez S, et al. Systemic the literature. Lupus 2002;11:722-9.
lupus erythematosus (SLE) in childhood: analysis of clinical and immunological [42] Garcia MA, Marcos JC, Marcos AI, Pons-Estel BA, Wojdyla D, Arturi A, et al. Male
findings in 34 patients and comparison with SLE characteristics in adults. Ann systemic lupus erythematosus in a Latin-American inception cohort of 1214
Rheum Dis 1998;57:456-9. patients. Lupus 2005;14:938-46.
[16] Hiraki LT, Benseler SM, Tyrrell PN, Hebert D, Harvey E, Silverman ED. Clinical and [43] Lee TP, Tang SJ, Wu MF, Song YC, Yu CL, Sun KH. Transgenic overexpression of
laboratory characteristics and long-term outcome of pediatric systemic lupus anti-double-stranded DNA autoantibody and activation of Toll-like receptor 4
erythematosus: a longitudinal study. J Pediatr 2008;152:550-6. in mice induce severe systemic lupus erythematosus syndromes. J Autoimmun
[17] Hoffman IE, Lauwerys BR, De Keyser F, Huizinga TW, Isenberg D, Cebecauer L, et al. 2010;35:358-67.
Juvenile-onset systemic lupus erythematosus: different clinical and serological [44] Berthier CC, Bethunaickan R, Gonzalez-Rivera T, Nair V, Ramanujam M, Zhang W,
pattern than adult-onset systemic lupus erythematosus. Ann Rheum Dis 2009;68: et al. Cross-species transcriptional network analysis defines shared inflammatory
412-5. responses in murine and human lupus nephritis. J Immunol 2012;189:988–1001.
[18] Mahmoud SS, Bazaraa HM, Lotfy HM, Abd-El-Aziz DM. Renal involvement in [45] Nguyen V, Luzina I, Rus H, Tegla C, Chen C, Rus V. IL-21 promotes lupus-like
childhood-onset systemic lupus erythematosus in Egypt. Rheumatol Int 2012;32: disease in chronic graft-versus-host disease through both CD4 T cell- and B
47-51. cell-intrinsic mechanisms. J Immunol 2012;189:1081-93.
[19] Boddaert J, Huong DL, Amoura Z, Wechsler B, Godeau P, Piette JC. Late-onset [46] Summers SA, Hoi A, Steinmetz OM, O'Sullivan KM, Ooi JD, Odobasic D, et al. TLR9
systemic lupus erythematosus: a personal series of 47 patients and pooled analysis and TLR4 are required for the development of autoimmunity and lupus nephritis
of 714 cases in the literature. Med (Baltimore) 2004;83:348-59. in pristane nephropathy. J Autoimmun 2010;35:291-8.
[20] Jacobsen S, Petersen J, Ullman S, Junker P, Voss A, Rasmussen JM, et al. A multicentre [47] Fletcher CA, Groom JR, Woehl B, Leung H, Mackay C, Mackay F. Development of
study of 513 Danish patients with systemic lupus erythematosus. I. Disease autoimmune nephritis in genetically asplenic and splenectomized BAFF transgenic
manifestations and analyses of clinical subsets. Clin Rheumatol 1998;17:468-77. mice. J Autoimmun 2011;36:125-34.
[21] Seligman VA, Lum RF, Olson JL, Li H, Criswell LA. Demographic differences in the [48] Baudino L, Yoshinobu K, Dunand-Sauthier I, Evans LH, Izui S. TLR-mediated
development of lupus nephritis: a retrospective analysis. Am J Med 2002;112: up-regulation of serum retroviral gp70 is controlled by the Sgp loci of lupus-
726-9. prone mice. J Autoimmun 2010;35:153-9.
[22] Alonso MD, Llorca J, Martinez-Vazquez F, Miranda-Filloy JA, Diaz de Teran T, [49] Koh AE, Njoroge SW, Feliu M, Cook A, Selig MK, Latchman YE, et al. The SLAM
Dierssen T, et al. Systemic lupus erythematosus in northwestern Spain: a 20-year family member CD48 (Slamf2) protects lupus-prone mice from autoimmune
epidemiologic study. Medicine (Baltimore) 2011;90:350-8. nephritis. J Autoimmun 2011;37:48-57.
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 189

[50] Potula HH, Morel L. Genetic variation at a Yin-Yang 1 response site regulates the [79] Xi J, Zhang GP, Qiao SL, Guo JQ, Wang XN, Yang YY, et al. Increased survival and
transcription of cyclin-dependent kinase inhibitor p18INK4C transcript in reduced renal injury in MRL/lpr mice treated with a human Fcgamma receptor II
lupus-prone mice. J Immunol 2012;188:4992-5002. (CD32) peptide. Immunology 2012;136:46-53.
[51] Perry DJ, Yin Y, Telarico T, Baker HV, Dozmorov I, Perl A, et al. Murine lupus [80] Werwitzke S, Trick D, Sondermann P, Kamino K, Schlegelberger B, Kniesch K,
susceptibility locus Sle1c2 mediates CD4+ T cell activation and maps to et al. Treatment of lupus-prone NZB/NZW F1 mice with recombinant soluble
estrogen-related receptor gamma. J Immunol 2012;189:793-803. Fc gamma receptor II (CD32). Ann Rheum Dis 2008;67:154-61.
[52] Baranowska-Daca E, Choi YJ, Barrios R, Nassar G, Suki WN, Truong LD. Nonlupus [81] Boulé MW, Broughton C, Mackay F, Akira S, Marshak-Rothstein A, Rifkin IR.
nephritides in patients with systemic lupus erythematosus: a comprehensive Toll-like receptor 9-dependent and -independent dendritic cell activation by
clinicopathologic study and review of the literature. Hum Pathol 2001;32: chromatin-immunoglobulin G complexes. J Exp Med 2004;199:1631-40.
1125-35. [82] Means TK, Latz E, Hayashi F, Murali MR, Golenbock DT, Luster AD. Human lupus
[53] Churg J, Bernstein J, Glassock RJ. Renal disease: classification and atlas of glomerular autoantibody–DNA complexes activate DCs through cooperation of CD32 and
disease. 2nd ed.New York: Igaku-Shoin; 1995. TLR9. J Clin Invest 2005;115:407-17.
[54] Weening JJ, D'Agati VD, Schwartz MM, Seshan SV, Alpers CE, Appel GB, et al. The [83] Cavallo T, Cameron WR, Lapenas D. Immunopathology of early and clinically
classification of glomerulonephritis in systemic lupus erythematosus revisited. silent lupus nephropathy. Am J Pathol 1977;87:1–18.
J Am Soc Nephrol 2004;15:241-50. [84] Gonzalez-Crespo MR, Lopez-Fernandez JI, Usera G, Poveda MJ, Gomez-Reino JJ.
[55] Najafi CC, Korbet SM, Lewis EJ, Schwartz MM, Reichlin M, Evans J. Significance of Outcome of silent lupus nephritis. Semin Arthritis Rheum 1996;26:468-76.
histologic patterns of glomerular injury upon long-term prognosis in severe [85] Wada Y, Ito S, Ueno M, Nakano M, Arakawa M, Gejyo F. Renal outcome and pre-
lupus glomerulonephritis. Kidney Int 2001;59:2156-63. dictors of clinical renal involvement in patients with silent lupus nephritis.
[56] Yokoyama H, Wada T, Hara A, Yamahana J, Nakaya I, Kobayashi M, et al. The Nephron Clin Pract 2004;98:c105-11.
outcome and a new ISN/RPS 2003 classification of lupus nephritis in Japanese. [86] Wakasugi D, Gono T, Kawaguchi Y, Hara M, Koseki Y, Katsumata Y, et al. Fre-
Kidney Int 2004;66:2382-8. quency of class III and IV nephritis in systemic lupus erythematosus without
[57] Hill GS, Delahousse M, Nochy D, Bariety J. Class IV-S versus class IV-G lupus clinical renal involvement: an analysis of predictive measures. J Rheumatol
nephritis: clinical and morphologic differences suggesting different pathogenesis. 2012;39:79-85.
Kidney Int 2005;68:2288-97. [87] Schiffer L, Sinha J, Wang X, Huang W, von Gersdorff G, Schiffer M, et al. Short
[58] Mittal B, Hurwitz S, Rennke H, Singh AK. New subcategories of class IV lupus term administration of costimulatory blockade and cyclophosphamide induces
nephritis: are there clinical, histologic, and outcome differences? Am J Kidney remission of systemic lupus erythematosus nephritis in NZB/W F1 mice by a
Dis 2004;44:1050-9. mechanism downstream of renal immune complex deposition. J Immunol
[59] Kim YG, Kim HW, Cho YM, Oh JS, Nah SS, Lee CK, et al. The difference between lupus 2003;171:489-97.
nephritis class IV-G and IV-S in Koreans: focus on the response to cyclophospha- [88] Jacob CO, Pricop L, Putterman C, Koss MN, Liu Y, Kollaros M, et al. Paucity of clinical
mide induction treatment. Rheumatology (Oxford) 2008;47:311-4. disease despite serological autoimmunity and kidney pathology in lupus-prone
[60] Hiramatsu N, Kuroiwa T, Ikeuchi H, Maeshima A, Kaneko Y, Hiromura K, et al. New Zealand mixed 2328 mice deficient in BAFF. J Immunol 2006;177:2671-80.
Revised classification of lupus nephritis is valuable in predicting renal outcome [89] Mannik M, Merrill CE, Stamps LD, Wener MH. Multiple autoantibodies form the
with an indication of the proportion of glomeruli affected by chronic lesions. glomerular immune deposits in patients with systemic lupus erythematosus.
Rheumatology (Oxford) 2008;47:702-7. J Rheumatol 2003;30:1495-504.
[61] Kojo S, Sada KE, Kobayashi M, Maruyama M, Maeshima Y, Sugiyama H, et al. [90] Mannik M, Wener MH. Deposition of antibodies to the collagen-like region of
Clinical usefulness of a prognostic score in histological analysis of renal biopsy C1q in renal glomeruli of patients with proliferative lupus glomerulonephritis.
in patients with lupus nephritis. J Rheumatol 2009;36:2218-23. Arthritis Rheum 1997;40:1504-11.
[62] Yu F, Tan Y, Wu LH, Zhu SN, Liu G, Zhao MH. Class IV-G and IV-S lupus nephritis [91] Reichlin M, Wolfson-Reichlin M. Evidence for the participation of anti-ribosomal
in Chinese patients: a large cohort study from a single center. Lupus 2009;18: P antibodies in lupus nephritis. Arthritis Rheum 1999;42:2728-9.
1073-81. [92] Krishnan C, Kaplan MH. Immunopathologic studies of systemic lupus
[63] Grootscholten C, Bajema IM, Florquin S, Steenbergen EJ, Peutz-Kootstra CJ, erythematosus. II. Antinuclear reaction of gamma-globulin eluted from homog-
Goldschmeding R, et al. Interobserver agreement of scoring of histopathological enates and isolated glomeruli of kidneys from patients with lupus nephritis.
characteristics and classification of lupus nephritis. Nephrol Dial Transplant J Clin Invest 1967;46:569-79.
2008;23:223-30. [93] Koffler D, Agnello V, Kimkel HG. Polynucleotide immune complexes in serum
[64] Schwartz MM, Korbet SM, Lewis EJ. The prognosis and pathogenesis of severe and glomeruli of patients with systemic lupus erythematosus. Am J Pathol
lupus glomerulonephritis. Nephrol Dial Transplant 2008;23:1298-306. 1974;74:109-24.
[65] Hoffmann MH, Trembleau S, Muller S, Steiner G. Nucleic acid-associated [94] Sasaki T, Hatakeyama A, Shibata S, Osaki H, Suzuki M, Horie K, et al. Heterogene-
autoantigens: pathogenic involvement and therapeutic potential. J Autoimmun ity of immune complex-derived anti-DNA antibodies associated with lupus ne-
2010;34:J178-206. phritis. Kidney Int 1991;39:746-53.
[66] Casciola-Rosen LA, Anhalt G, Rosen A. Autoantigens targeted in systemic lupus [95] Arbuckle MR, McClain MT, Rubertone MV, Scofield RH, Dennis GJ, James JA, et al.
erythematosus are clustered in two populations of surface structures on apopto- Development of autoantibodies before the clinical onset of systemic lupus
tic keratinocytes. J Exp Med 1994;179:1317-30. erythematosus. N Engl J Med 2003;349:1526-33.
[67] Gaipl US, Munoz LE, Grossmayer G, Lauber K, Franz S, Sarter K, et al. Clearance [96] Isenberg DA, Manson JJ, Ehrenstein MR, Rahman A. Fifty years of anti-ds DNA
deficiency and systemic lupus erythematosus (SLE). J Autoimmun 2007;28: antibodies: are we approaching journey's end? Rheumatology (Oxford) 2007;46:
114-21. 1052-6.
[68] Herrmann M, Voll RE, Zoller OM, Hagenhofer M, Ponner BB, Kalden JR. Impaired [97] Smykal-Jankowiak K, Niemir ZI, Polcyn-Adamczak M. Do circulating antibodies
phagocytosis of apoptotic cell material by monocyte-derived macrophages from against C1q reflect the activity of lupus nephritis? Pol Arch Med Wewn
patients with systemic lupus erythematosus. Arthritis Rheum 1998;41:1241-50. 2011;121:287-95.
[69] Burlingame RW, Boey ML, Starkebaum G, Rubin RL. The central role of [98] Das L, Brunner HI. Biomarkers for renal disease in childhood. Curr Rheumatol
chromatin in autoimmune responses to histones and DNA in systemic lupus Rep 2009;11:218-25.
erythematosus. J Clin Invest 1994;94:184-92. [99] Ehrenstein MR, Katz DR, Griffiths MH, Papadaki L, Winkler TH, Kalden JR, et al.
[70] Urbonaviciute V, Furnrohr BG, Meister S, Munoz L, Heyder P, De Marchis F, et al. Human IgG anti-DNA antibodies deposit in kidneys and induce proteinuria in
Induction of inflammatory and immune responses by HMGB1-nucleosome com- SCID mice. Kidney Int 1995;48:705-11.
plexes: implications for the pathogenesis of SLE. J Exp Med 2008;205:3007-18. [100] Vlahakos DV, Foster MH, Adams S, Katz M, Ucci AA, Barrett KJ, et al. Anti-DNA
[71] Abdulahad DA, Westra J, Bijzet J, Limburg PC, Kallenberg CG, Bijl M. High mobil- antibodies form immune deposits at distinct glomerular and vascular sites. Kidney
ity group box 1 (HMGB1) and anti-HMGB1 antibodies and their relation to Int 1992;41:1690-700.
disease characteristics in systemic lupus erythematosus. Arthritis Res Ther [101] Mostoslavsky G, Fischel R, Yachimovich N, Yarkoni Y, Rosenmann E, Monestier
2011;13:R71. M, et al. Lupus anti-DNA autoantibodies cross-react with a glomerular structural
[72] Zickert A, Palmblad K, Sundelin B, Chavan S, Tracey KJ, Bruchfeld A, et al. Renal protein: a case for tissue injury by molecular mimicry. Eur J Immunol 2001;31:
expression and serum levels of high mobility group box 1 protein in lupus ne- 1221-7.
phritis. Arthritis Res Ther 2012;14:R36. [102] Deocharan B, Qing X, Lichauco J, Putterman C. Alpha-actinin is a cross-reactive
[73] Urbonaviciute V, Voll RE. High-mobility group box 1 represents a potential renal target for pathogenic anti-DNA antibodies. J Immunol 2002;168:3072-8.
marker of disease activity and novel therapeutic target in systemic lupus [103] Renaudineau Y, Croquefer S, Jousse S, Renaudineau E, Devauchelle V, Gueguen P,
erythematosus. J Intern Med 2011;270:309-18. et al. Association of alpha-actinin-binding anti-double-stranded DNA antibodies
[74] Botto M, Walport MJ. C1q, autoimmunity and apoptosis. Immunobiology with lupus nephritis. Arthritis Rheum 2006;54:2523-32.
2002;205:395-406. [104] Rekvig OP, Putterman C, Casu C, Gao HX, Ghirardello A, Mortensen ES, et al. Autoanti-
[75] Radanova M, Vasilev V, Deliyska B, Kishore U, Ikonomov V, Ivanova D. Anti-C1q auto- bodies in lupus: culprits or passive bystanders? Autoimmun Rev 2012;11:596-603.
antibodies specific against the globular domain of the C1qB-chain from patient with [105] van Bavel CC, Fenton KA, Rekvig OP, van der Vlag J, Berden JH. Glomerular targets
lupus nephritis inhibit C1q binding to IgG and CRP. Immunobiology 2012;217:684-91. of nephritogenic autoantibodies in systemic lupus erythematosus. Arthritis
[76] Nowling TK, Gilkeson GS. Mechanisms of tissue injury in lupus nephritis. Arthri- Rheum 2008;58:1892-9.
tis Res Ther 2011;13:250. [106] Zhao Z, Deocharan B, Scherer PE, Ozelius LJ, Putterman C. Differential binding of
[77] Sato N, Ohsawa I, Nagamachi S, Ishii M, Kusaba G, Inoshita H, et al. Significance of cross-reactive anti-DNA antibodies to mesangial cells: the role of alpha-actinin.
glomerular activation of the alternative pathway and lectin pathway in lupus ne- J Immunol 2006;176:7704-14.
phritis. Lupus 2011;20:1378-86. [107] Deocharan B, Zhou Z, Antar K, Siconolfi-Baez L, Angeletti RH, Hardin J, et al. Alpha-
[78] Clynes R, Dumitru C, Ravetch JV. Uncoupling of immune complex formation and actinin immunization elicits anti-chromatin autoimmunity in nonautoimmune
kidney damage in autoimmune glomerulonephritis. Science 1998;279:1052-4. mice. J Immunol 2007;179:1313-21.
190 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

[108] Kalaaji M, Fenton KA, Mortensen ES, Olsen R, Sturfelt G, Alm P, et al. Glomerular [135] Feng X, Wu H, Grossman JM, Hanvivadhanakul P, FitzGerald JD, Park GS, et al.
apoptotic nucleosomes are central target structures for nephritogenic antibodies Association of increased interferon-inducible gene expression with disease
in human SLE nephritis. Kidney Int 2007;71:664-72. activity and lupus nephritis in patients with systemic lupus erythematosus.
[109] Kalaaji M, Mortensen E, Jorgensen L, Olsen R, Rekvig OP. Nephritogenic lupus Arthritis Rheum 2006;54:2951-62.
antibodies recognize glomerular basement membrane-associated chromatin [136] Kirou KA, Lee C, George S, Louca K, Peterson MG, Crow MK. Activation of the
fragments released from apoptotic intraglomerular cells. Am J Pathol 2006;168: interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus
1779-92. patients with distinct serologic features and active disease. Arthritis Rheum
[110] Mjelle JE, Rekvig OP, Van Der Vlag J, Fenton KA. Nephritogenic antibodies bind in 2005;52:1491-503.
glomeruli through interaction with exposed chromatin fragments and not with [137] Landolt-Marticorena C, Bonventi G, Lubovich A, Ferguson C, Unnithan T, Su J,
renal cross-reactive antigens. Autoimmunity 2011;44:373-83. et al. Lack of association between the interferon-alpha signature and longitudi-
[111] Waters ST, McDuffie M, Bagavant H, Deshmukh US, Gaskin F, Jiang C, et al. Break- nal changes in disease activity in systemic lupus erythematosus. Ann Rheum
ing tolerance to double stranded DNA, nucleosome, and other nuclear antigens is Dis 2009;68:1440-6.
not required for the pathogenesis of lupus glomerulonephritis. J Exp Med [138] Petri M, Singh S, Tesfasyone H, Dedrick R, Fry K, Lal P, et al. Longitudinal expres-
2004;199:255-64. sion of type I interferon responsive genes in systemic lupus erythematosus.
[112] Christensen SR, Shupe J, Nickerson K, Kashgarian M, Flavell RA, Shlomchik MJ. Lupus 2009;18:980-9.
Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing [139] Weckerle CE, Franek BS, Kelly JA, Kumabe M, Mikolaitis RA, Green SL, et al. Net-
inflammatory and regulatory roles in a murine model of lupus. Immunity work analysis of associations between serum interferon-alpha activity, autoanti-
2006;25:417-28. bodies, and clinical features in systemic lupus erythematosus. Arthritis Rheum
[113] Sinico RA, Radice A, Ikehata M, Giammarresi G, Corace C, Arrigo G, et al. Anti-C1q 2011;63:1044-53.
autoantibodies in lupus nephritis: prevalence and clinical significance. Ann N Y [140] Kariuki SN, Kirou KA, MacDermott EJ, Barillas-Arias L, Crow MK, Niewold TB.
Acad Sci 2005;1050:193-200. Cutting edge: autoimmune disease risk variant of STAT4 confers increased sen-
[114] Mok CC, Ho LY, Leung HW, Wong LG. Performance of anti-C1q, antinucleosome, sitivity to IFN-alpha in lupus patients in vivo. J Immunol 2009;182:34-8.
and anti-dsDNA antibodies for detecting concurrent disease activity of systemic [141] Fairhurst AM, Xie C, Fu Y, Wang A, Boudreaux C, Zhou XJ, et al. Type I interferons
lupus erythematosus. Transl Res 2010;156:320-5. produced by resident renal cells may promote end-organ disease in autoantibody-
[115] Cortés-Hernández J, Ordi-Ros J, Labrador M, Segarra A, Tovar JL, Balada E, et al. Predic- mediated glomerulonephritis. J Immunol 2009;183:6831-8.
tors of poor renal outcome in patients with lupus nephritis treated with combined [142] Agrawal H, Jacob N, Carreras E, Bajana S, Putterman C, Turner S, et al. Deficiency
pulses of cyclophosphamide and methylprednisolone. Lupus 2003;12:287-96. of type I IFN receptor in lupus-prone New Zealand mixed 2328 mice decreases
[116] Julkunen H, Ekblom-Kullberg S, Miettinen A. Nonrenal and renal activity of sys- dendritic cell numbers and activation and protects from disease. J Immunol
temic lupus erythematosus: a comparison of two anti-C1q and five anti-dsDNA 2009;183:6021-9.
assays and complement C3 and C4. Rheumatol Int 2012;32:2445-51. [143] Santiago-Raber ML, Baccala R, Haraldsson KM, Choubey D, Stewart TA, Kono DH,
[117] Trendelenburg M, Lopez-Trascasa M, Potlukova E, Moll S, Regenass S, Fremeaux- et al. Type-I interferon receptor deficiency reduces lupus-like disease in NZB
Bacchi V, et al. High prevalence of anti-C1q antibodies in biopsy-proven active mice. J Exp Med 2003;197:777-88.
lupus nephritis. Nephrol Dial Transplant 2006;21:3115-21. [144] Braun D, Geraldes P, Demengeot J. Type I interferon controls the onset and severity
[118] Marto N, Bertolaccini ML, Calabuig E, Hughes GR, Khamashta MA. Anti-C1q antibodies of autoimmune manifestations in lpr mice. J Autoimmun 2003;20:15-25.
in nephritis: correlation between titres and renal disease activity and positive predic- [145] Hron JD, Peng SL. Type I IFN protects against murine lupus. J Immunol 2004;173:
tive value in systemic lupus erythematosus. Ann Rheum Dis 2005;64:444-8. 2134-42.
[119] Cai X, Yang X, Lian F, Lin X, Liang M, Li J, et al. Correlation between serum anti-C1q [146] Mathian A, Gallegos M, Pascual V, Banchereau J, Koutouzov S. Interferon-alpha
antibody levels and renal pathological characteristics and prognostic significance of induces unabated production of short-lived plasma cells in pre-autoimmune
anti-C1q antibody in lupus nephritis. J Rheumatol 2010;37:759-65. lupus-prone (NZBxNZW)F1 mice but not in BALB/c mice. Eur J Immunol
[120] Coremans IE, Spronk PE, Bootsma H, Daha MR, van der Voort EA, Kater L, et al. 2011;41:863-72.
Changes in antibodies to C1q predict renal relapses in systemic lupus [147] Liu Z, Bethunaickan R, Huang W, Lodhi U, Solano I, Madaio MP, et al. Interferon-
erythematosus. Am J Kidney Dis 1995;26:595-601. alpha accelerates murine systemic lupus erythematosus in a T cell-dependent
[121] Moroni G, Radice A, Giammarresi G, Quaglini S, Gallelli B, Leoni A, et al. Are lab- manner. Arthritis Rheum 2011;63:219-29.
oratory tests useful for monitoring the activity of lupus nephritis? A 6-year pro- [148] Fairhurst AM, Mathian A, Connolly JE, Wang A, Gray HF, George TA, et al. Systemic
spective study in a cohort of 228 patients with lupus nephritis. Ann Rheum Dis IFN-alpha drives kidney nephritis in B6.Sle123 mice. Eur J Immunol 2008;38:
2009;68:234-7. 1948-60.
[122] Trendelenburg M, Marfurt J, Gerber I, Tyndall A, Schifferli JA. Lack of occurrence [149] Liu Z, Bethunaickan R, Huang W, Ramanujam M, Madaio MP, Davidson A.
of severe lupus nephritis among anti-C1q autoantibody-negative patients. Ar- IFN-alpha confers resistance of systemic lupus erythematosus nephritis to therapy
thritis Rheum 1999;42:187-8. in NZB/W F1 mice. J Immunol 2011;187:1506-13.
[123] Meyer OC, Nicaise-Roland P, Cadoudal N, Grootenboer-Mignot S, Palazzo E, [150] Lövgren T, Eloranta ML, Båve U, Alm GV, Rönnblom L. Induction of interferon-alpha
Hayem G, et al. Anti-C1q antibodies antedate patent active glomerulonephritis production in plasmacytoid dendritic cells by immune complexes containing
in patients with systemic lupus erythematosus. Arthritis Res Ther 2009;11:R87. nucleic acid released by necrotic or late apoptotic cells and lupus IgG. Arthritis
[124] Katsumata Y, Miyake K, Kawaguchi Y, Okamoto Y, Kawamoto M, Gono T, et al. Rheum 2004;50:1861-72.
Anti-C1q antibodies are associated with systemic lupus erythematosus global [151] Barrat FJ, Meeker T, Gregorio J, Chan JH, Uematsu S, Akira S, et al. Nucleic acids of
activity but not specifically with nephritis: a controlled study of 126 consecutive mammalian origin can act as endogenous ligands for Toll-like receptors and may
patients. Arthritis Rheum 2011;63:2436-44. promote systemic lupus erythematosus. J Exp Med 2005;202:1131-9.
[125] Trouw LA, Groeneveld TW, Seelen MA, Duijs JM, Bajema IM, Prins FA, et al. [152] Christensen SR, Shlomchik MJ. Regulation of lupus-related autoantibody production
Anti-C1q autoantibodies deposit in glomeruli but are only pathogenic in combi- and clinical disease by Toll-like receptors. Semin Immunol 2007;19:11-23.
nation with glomerular C1q-containing immune complexes. J Clin Invest [153] Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, et al. Toll-like receptor
2004;114:679-88. 9-dependent activation by DNA-containing immune complexes is mediated by
[126] O'Flynn J, Flierman R, van der Pol P, Rops A, Satchell SC, Mathieson PW, et al. Nucle- HMGB1 and RAGE. Nat Immunol 2007;8:487-96.
osomes and C1q bound to glomerular endothelial cells serve as targets for autoan- [154] Lood C, Gullstrand B, Truedsson L, Olin AI, Alm GV, Rönnblom L, et al. C1q inhibits
tibodies and determine complement activation. Mol Immunol 2011;49:75-83. immune complex-induced interferon-alpha production in plasmacytoid dendritic
[127] Yang XW, Tan Y, Yu F, Zhao MH. Combination of anti-C1q and anti-dsDNA anti- cells: a novel link between C1q deficiency and systemic lupus erythematosus
bodies is associated with higher renal disease activity and predicts renal progno- pathogenesis. Arthritis Rheum 2009;60:3081-90.
sis of patients with lupus nephritis. Nephrol Dial Transplant 2012;27:3552-9. [155] Santer DM, Hall BE, George TC, Tangsombatvisit S, Liu CL, Arkwright PD, et al.
[128] Preble OT, Black RJ, Friedman RM, Klippel JH, Vilcek J. Systemic lupus C1q deficiency leads to the defective suppression of IFN-alpha in response to
erythematosus: presence in human serum of an unusual acid-labile leukocyte nucleoprotein containing immune complexes. J Immunol 2010;185:4738-49.
interferon. Science 1982;216:429-31. [156] Schreibelt G, Tel J, Sliepen KH, Benitez-Ribas D, Figdor CG, Adema GJ, et al.
[129] Kim T, Kanayama Y, Negoro N, Okamura M, Takeda T, Inoue T. Serum levels of in- Toll-like receptor expression and function in human dendritic cell subsets:
terferons in patients with systemic lupus erythematosus. Clin Exp Immunol implications for dendritic cell-based anti-cancer immunotherapy. Cancer
1987;70:562-9. Immunol Immunother 2010;59:1573-82.
[130] Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, et al. [157] Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, et al. Netting neu-
Interferon-inducible gene expression signature in peripheral blood cells of pa- trophils are major inducers of type I IFN production in pediatric systemic lupus
tients with severe lupus. Proc Natl Acad Sci U S A 2003;100:2610-5. erythematosus. Sci Transl Med 2011;3:73ra20.
[131] Bennett L, Palucka AK, Arce E, Cantrell V, Borvak J, Banchereau J, et al. Interferon [158] Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, et al. Neutrophils
and granulopoiesis signatures in systemic lupus erythematosus blood. J Exp Med activate plasmacytoid dendritic cells by releasing self-DNA–peptide complexes in
2003;197:711-23. systemic lupus erythematosus. Sci Transl Med 2011;3:73ra19.
[132] Hua J, Kirou K, Lee C, Crow MK. Functional assay of type I interferon in systemic [159] Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, et al. Impair-
lupus erythematosus plasma and association with anti-RNA binding protein au- ment of neutrophil extracellular trap degradation is associated with lupus nephri-
toantibodies. Arthritis Rheum 2006;54:1906-16. tis. Proc Natl Acad Sci U S A 2010;107:9813-8.
[133] Blanco P, Palucka AK, Gill M, Pascual V, Banchereau J. Induction of dendritic cell differ- [160] Guiducci C, Gong M, Xu Z, Gill M, Chaussabel D, Meeker T, et al. TLR recognition of self
entiation by IFN-alpha in systemic lupus erythematosus. Science 2001;294:1540-3. nucleic acids hampers glucocorticoid activity in lupus. Nature 2010;465:937-41.
[134] Nikpour M, Dempsey AA, Urowitz MB, Gladman DD, Barnes DA. Association of a [161] Anders HJ, Vielhauer V, Eis V, Linde Y, Kretzler M, Perez de Lema G, et al. Activa-
gene expression profile from whole blood with disease activity in systemic lupus tion of toll-like receptor-9 induces progression of renal disease in MRL-Fas(lpr)
erythaematosus. Ann Rheum Dis 2008;67:1069-75. mice. FASEB J 2004;18:534-6.
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 191

[162] Pawar RD, Patole PS, Ellwart A, Lech M, Segerer S, Schlondorff D, et al. Ligands to [191] Lech M, Weidenbusch M, Kulkarni OP, Ryu M, Darisipudi MN, Susanti HE, et al.
nucleic acid-specific toll-like receptors and the onset of lupus nephritis. J Am Soc IRF4 deficiency abrogates lupus nephritis despite enhancing systemic cytokine
Nephrol 2006;17:3365-73. production. J Am Soc Nephrol 2011;22:1443-52.
[163] Pawar RD, Patole PS, Zecher D, Segerer S, Kretzler M, Schlondorff D, et al. [192] Meroni PL, Shoenfeld Y. Systemic lupus erythematosus and the SLE galaxy.
Toll-like receptor-7 modulates immune complex glomerulonephritis. J Am Soc Autoimmun Rev 2010;10:1-2.
Nephrol 2006;17:141-9. [193] Ramos PS, Brown EE, Kimberly RP, Langefeld CD. Genetic factors predisposing to
[164] Conti F, Spinelli FR, Alessandri C, Valesini G. Toll-like receptors and lupus nephri- systemic lupus erythematosus and lupus nephritis. Semin Nephrol 2010;30:
tis. Clin Rev Allergy Immunol 2011;40:192-8. 164-76.
[165] Nickerson KM, Christensen SR, Shupe J, Kashgarian M, Kim D, Elkon K, et al. TLR9 [194] Marchini M, Antonioli R, Lleo A, Barili M, Caronni M, Origgi L, et al. HLA class II
regulates TLR7- and MyD88-dependent autoantibody production and disease in antigens associated with lupus nephritis in Italian SLE patients. Hum Immunol
a murine model of lupus. J Immunol 2010;184:1840-8. 2003;64:462-8.
[166] Papadimitraki ED, Tzardi M, Bertsias G, Sotsiou E, Boumpas DT. Glomerular ex- [195] Li LH, Yuan H, Pan HF, Li WX, Li XP, Ye DQ. Role of the Fcgamma receptor
pression of toll-like receptor-9 in lupus nephritis but not in normal kidneys: im- IIIA-V/F158 polymorphism in susceptibility to systemic lupus erythematosus
plications for the amplification of the inflammatory response. Lupus 2009;18: and lupus nephritis: a meta-analysis. Scand J Rheumatol 2010;39:148-54.
831-5. [196] Yuan H, Ni JD, Pan HF, Li LH, Feng JB, Ye DQ. Lack of association of
[167] Frieri M, Samih MA, Dzhindzhikhashvili M, Liu H, Balsam L, Rubinstein S. FcgammaRIIIb polymorphisms with systemic lupus erythematosus: a meta-
Toll-like receptor 9 and vascular endothelial growth factor levels in human kid- analysis. Rheumatol Int 2011;31:1017-21.
neys from lupus nephritis patients. J Nephrol 2012;0. [197] Lee YH, Ji JD, Song GG. Fcgamma receptor IIB and IIIB polymorphisms and suscepti-
[168] Benigni A, Caroli C, Longaretti L, Gagliardini E, Zoja C, Galbusera M, et al. Involve- bility to systemic lupus erythematosus and lupus nephritis: a meta-analysis. Lupus
ment of renal tubular Toll-like receptor 9 in the development of tubulointerstitial 2009;18:727-34.
injury in systemic lupus. Arthritis Rheum 2007;56:1569-78. [198] Taylor KE, Remmers EF, Lee AT, Ortmann WA, Plenge RM, Tian C, et al. Specificity
[169] Chan OT, Hannum LG, Haberman AM, Madaio MP, Shlomchik MJ. A novel mouse of the STAT4 genetic association for severe disease manifestations of systemic
with B cells but lacking serum antibody reveals an antibody-independent role lupus erythematosus. PLoS Genet 2008;4:e1000084.
for B cells in murine lupus. J Exp Med 1999;189:1639-48. [199] Sigurdsson S, Nordmark G, Garnier S, Grundberg E, Kwan T, Nilsson O, et al. A
[170] Steinmetz OM, Velden J, Kneissler U, Marx M, Klein A, Helmchen U, et al. Analy- risk haplotype of STAT4 for systemic lupus erythematosus is over-expressed,
sis and classification of B-cell infiltrates in lupus and ANCA-associated nephritis. correlates with anti-dsDNA and shows additive effects with two risk alleles of
Kidney Int 2008;74:448-57. IRF5. Hum Mol Genet 2008;17:2868-76.
[171] Chang A, Henderson SG, Brandt D, Liu N, Guttikonda R, Hsieh C, et al. In situ B [200] Vuong MT, Gunnarsson I, Lundberg S, Svenungsson E, Wramner L, Fernstrom A,
cell-mediated immune responses and tubulointerstitial inflammation in et al. Genetic risk factors in lupus nephritis and IgA nephropathy—no support of
human lupus nephritis. J Immunol 2011;186:1849-60. an overlap. PLoS One 2010;5:e10559.
[172] Espeli M, Bokers S, Giannico G, Dickinson HA, Bardsley V, Fogo AB, et al. Local [201] Kawasaki A, Ito I, Hikami K, Ohashi J, Hayashi T, Goto D, et al. Role of STAT4 poly-
renal autoantibody production in lupus nephritis. J Am Soc Nephrol 2011;22: morphisms in systemic lupus erythematosus in a Japanese population: a case–
296-305. control association study of the STAT1–STAT4 region. Arthritis Res Ther
[173] Couzi L, Merville P, Deminiere C, Moreau JF, Combe C, Pellegrin JL, et al. Predom- 2008;10:R113.
inance of CD8+ T lymphocytes among periglomerular infiltrating cells and link [202] Li P, Cao C, Luan H, Li C, Hu C, Zhang S, et al. Association of genetic variations in
to the prognosis of class III and class IV lupus nephritis. Arthritis Rheum the STAT4 and IRF7/KIAA1542 regions with systemic lupus erythematosus in a
2007;56:2362-70. Northern Han Chinese population. Hum Immunol 2011;72:249-55.
[174] Alexopoulos E, Seron D, Hartley RB, Cameron JS. Lupus nephritis: correlation of [203] Abelson AK, Delgado-Vega AM, Kozyrev SV, Sanchez E, Velazquez-Cruz R,
interstitial cells with glomerular function. Kidney Int 1990;37:100-9. Eriksson N, et al. STAT4 associates with systemic lupus erythematosus through
[175] Boucher A, Droz D, Adafer E, Noel LH. Characterization of mononuclear cell sub- two independent effects that correlate with gene expression and act additively
sets in renal cellular interstitial infiltrates. Kidney Int 1986;29:1043-9. with IRF5 to increase risk. Ann Rheum Dis 2009;68:1746-53.
[176] D'Agati VD, Appel GB, Estes D, Knowles II DM, Pirani CL. Monoclonal antibody [204] Jacob CO, Zang S, Li L, Ciobanu V, Quismorio F, Mizutani A, et al. Pivotal role of
identification of infiltrating mononuclear leukocytes in lupus nephritis. Kidney Stat4 and Stat6 in the pathogenesis of the lupus-like disease in the New Zealand
Int 1986;30:573-81. mixed 2328 mice. J Immunol 2003;171:1564-71.
[177] Winchester R, Wiesendanger M, Zhang HZ, Steshenko V, Peterson K, [205] Singh RR, Saxena V, Zang S, Li L, Finkelman FD, Witte DP, et al. Differential con-
Geraldino-Pardilla L, et al. Immunologic characteristics of intrarenal T cells: traf- tribution of IL-4 and STAT6 vs STAT4 to the development of lupus nephritis. J
ficking of expanded CD8+ T cell beta-chain clonotypes in progressive lupus ne- Immunol 2003;170:4818-25.
phritis. Arthritis Rheum 2012;64:1589-600. [206] Xu Z, Duan B, Croker BP, Morel L. STAT4 deficiency reduces autoantibody pro-
[178] Yu F, Wu LH, Tan Y, Li LH, Wang CL, Wang WK, et al. Tubulointerstitial lesions of duction and glomerulonephritis in a mouse model of lupus. Clin Immunol
patients with lupus nephritis classified by the 2003 International Society of Ne- 2006;120:189-98.
phrology and Renal Pathology Society system. Kidney Int 2010;77:820-9. [207] Han JW, Zheng HF, Cui Y, Sun LD, Ye DQ, Hu Z, et al. Genome-wide association
[179] Hsieh C, Chang A, Brandt D, Guttikonda R, Utset TO, Clark MR. Predicting out- study in a Chinese Han population identifies nine new susceptibility loci for sys-
comes of lupus nephritis with tubulointerstitial inflammation and scarring. Ar- temic lupus erythematosus. Nat Genet 2009;41:1234-7.
thritis Care Res 2011;63:865-74. [208] He CF, Liu YS, Cheng YL, Gao JP, Pan TM, Han JW, et al. TNIP1, SLC15A4, ETS1,
[180] Dolff S, Abdulahad WH, van Dijk MC, Limburg PC, Kallenberg CG, Bijl M. Urinary RasGRP3 and IKZF1 are associated with clinical features of systemic lupus
T cells in active lupus nephritis show an effector memory phenotype. Ann erythematosus in a Chinese Han population. Lupus 2010;19:1181-6.
Rheum Dis 2010;69:2034-41. [209] Hu W, Ren H. A meta-analysis of the association of IRF5 polymorphism with sys-
[181] Massengill SF, Goodenow MM, Sleasman JW. SLE nephritis is associated with an temic lupus erythematosus. Int J Immunogenet 2011;38:411-7.
oligoclonal expansion of intrarenal T cells. Am J Kidney Dis 1998;31:418-26. [210] Harley JB, Alarcon-Riquelme ME, Criswell LA, Jacob CO, Kimberly RP, Moser KL,
[182] Murata H, Matsumura R, Koyama A, Sugiyama T, Sueishi M, Shibuya K, et al. T et al. Genome-wide association scan in women with systemic lupus
cell receptor repertoire of T cells in the kidneys of patients with lupus nephritis. erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and
Arthritis Rheum 2002;46:2141-7. other loci. Nat Genet 2008;40:204-10.
[183] Bagavant H, Deshmukh US, Wang H, Ly T, Fu SM. Role for nephritogenic T cells in [211] Durán-Barragán S, McGwin Jr G, Vilá LM, Reveille JD, Alarcón GS. Angiotensin-
lupus glomerulonephritis: progression to renal failure is accompanied by T cell converting enzyme inhibitors delay the occurrence of renal involvement and
activation and expansion in regional lymph nodes. J Immunol 2006;177: are associated with a decreased risk of disease activity in patients with systemic
8258-65. lupus erythematosus—results from LUMINA (LIX): a multiethnic US cohort.
[184] Fiore N, Castellano G, Blasi A, Capobianco C, Loverre A, Montinaro V, et al. Imma- Rheumatology (Oxford) 2008;47:1093-6.
ture myeloid and plasmacytoid dendritic cells infiltrate renal tubulointerstitium [212] Uchida K, Nitta K. Recent advances in the treatment of lupus nephritis. Clin Exp
in patients with lupus nephritis. Mol Immunol 2008;45:259-65. Nephrol 2011.
[185] Tucci M, Quatraro C, Lombardi L, Pellegrino C, Dammacco F, Silvestris F. Glomer- [213] Hahn BH, McMahon MA, Wilkinson A, Wallace WD, Daikh DI, Fitzgerald JD, et al.
ular accumulation of plasmacytoid dendritic cells in active lupus nephritis: role American College of Rheumatology guidelines for screening, treatment, and
of interleukin-18. Arthritis Rheum 2008;58:251-62. management of lupus nephritis. Arthritis Care Res 2012;64:797-808.
[186] Pappu R, Ramirez-Carrozzi V, Sambandam A. The interleukin-17 cytokine family: [214] Pollak VE, Pirani CL, Kark RM. Effect of large doses of prednisone on the renal le-
critical players in host defence and inflammatory diseases. Immunology sions and life span of patients with lupus glomerulonephritis. J Lab Clin Med
2011;134:8–16. 1961;57:495-511.
[187] Wong CK, Lit LC, Tam LS, Li EK, Wong PT, Lam CW. Hyperproduction of IL-23 and [215] Austin III HA, Klippel JH, Balow JE, le Riche NG, Steinberg AD, Plotz PH, et al.
IL-17 in patients with systemic lupus erythematosus: implications for Th17- Therapy of lupus nephritis. Controlled trial of prednisone and cytotoxic drugs.
mediated inflammation in auto-immunity. Clin Immunol 2008;127:385-93. N Engl J Med 1986;314:614-9.
[188] Kitching AR, Holdsworth SR. The emergence of TH17 cells as effectors of renal in- [216] Gourley MF, Austin III HA, Scott D, Yarboro CH, Vaughan EM, Muir J, et al. Meth-
jury. J Am Soc Nephrol 2011;22:235-8. ylprednisolone and cyclophosphamide, alone or in combination, in patients with
[189] Edelbauer M, Kshirsagar S, Riedl M, Billing H, Tonshoff B, Haffner D, et al. Activity lupus nephritis. A randomized, controlled trial. Ann Intern Med 1996;125:
of childhood lupus nephritis is linked to altered T cell and cytokine homeostasis. 549-57.
J Clin Immunol 2012. [217] Illei GG, Austin HA, Crane M, Collins L, Gourley MF, Yarboro CH, et al. Combina-
[190] Crispín JC, Oukka M, Bayliss G, Cohen RA, Van Beek CA, Stillman IE, et al. Expand- tion therapy with pulse cyclophosphamide plus pulse methylprednisolone im-
ed double negative T cells in patients with systemic lupus erythematosus pro- proves long-term renal outcome without adding toxicity in patients with lupus
duce IL-17 and infiltrate the kidneys. J Immunol 2008;181:8761-6. nephritis. Ann Intern Med 2001;135:248-57.
192 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

[218] Boumpas DT, Austin III HA, Vaughn EM, Klippel JH, Steinberg AD, Yarboro CH, [244] Rivera TL, Belmont HM, Malani S, Latorre M, Benton L, Weisstuch J, et al. Current
et al. Controlled trial of pulse methylprednisolone versus two regimens of therapies for lupus nephritis in an ethnically heterogeneous cohort. J Rheumatol
pulse cyclophosphamide in severe lupus nephritis. Lancet 1992;340:741-5. 2009;36:298-305.
[219] Gordon C, Jayne D, Pusey C, Adu D, Amoura Z, Aringer M, et al. European consen- [245] Isenberg D, Appel GB, Contreras G, Dooley MA, Ginzler EM, Jayne D, et al. Influ-
sus statement on the terminology used in the management of lupus glomerulo- ence of race/ethnicity on response to lupus nephritis treatment: the ALMS study.
nephritis. Lupus 2009;18:257-63. Rheumatology (Oxford) 2010;49:128-40.
[220] Renal Disease Subcommittee of the American College of Rheumatology Ad Hoc [246] Punyawudho B, Lertdumrongluk P, Somparn P, Kittanamongkolchai W, Traitanon O,
Committee on Systemic Lupus Erythematosus Response Criteria. The American Avihingsanon Y, et al. Population pharmacokinetics of mycophenolate mofetil in
College of Rheumatology response criteria for proliferative and membranous Thai lupus nephritis patients. Int J Clin Pharmacol Ther 2012;50:272-80.
renal disease in systemic lupus erythematosus clinical trials. Arthritis Rheum [247] Chan TM, Tse KC, Tang CS, Mok MY, Li FK. Long-term study of mycophenolate
2006;54:421-32. mofetil as continuous induction and maintenance treatment for diffuse prolifer-
[221] Korbet SM, Lewis EJ, Schwartz MM, Reichlin M, Evans J, Rohde RD. Factors pre- ative lupus nephritis. J Am Soc Nephrol 2005;16:1076-84.
dictive of outcome in severe lupus nephritis. Lupus Nephritis Collaborative [248] Weng MY, Weng CT, Liu MF. The efficacy of low-dose mycophenolate mofetil for
Study Group. Am J Kidney Dis 2000;35:904-14. treatment of lupus nephritis in Taiwanese patients with systemic lupus
[222] Ioannidis JP, Boki KA, Katsorida ME, Drosos AA, Skopouli FN, Boletis JN, et al. erythematosus. Clin Rheumatol 2010;29:771-5.
Remission, relapse, and re-remission of proliferative lupus nephritis treated [249] Wang J, Hu W, Xie H, Zhang H, Chen H, Zeng C, et al. Induction therapies for class IV
with cyclophosphamide. Kidney Int 2000;57:258-64. lupus nephritis with non-inflammatory necrotizing vasculopathy: mycophenolate
[223] So MW, Koo BS, Kim YG, Lee CK, Yoo B. Predictive value of remission status after mofetil or intravenous cyclophosphamide. Lupus 2007;16:707-12.
6 months induction therapy in patients with proliferative lupus nephritis: a ret- [250] Tang Z, Yang G, Yu C, Yu Y, Wang J, Hu W, et al. Effects of mycophenolate mofetil
rospective analysis. Clin Rheumatol 2011;30:1399-405. for patients with crescentic lupus nephritis. Nephrology (Carlton) 2008;13:
[224] Contreras G, Pardo V, Leclercq B, Lenz O, Tozman E, O'Nan P, et al. Sequential 702-7.
therapies for proliferative lupus nephritis. N Engl J Med 2004;350:971-80. [251] Wilson EC, Jayne DR, Dellow E, Fordham RJ. The cost-effectiveness of mycophenolate
[225] Houssiau FA, Vasconcelos C, D'Cruz D, Sebastiani GD, Garrido Ed Ede R, Danieli mofetil as firstline therapy in active lupus nephritis. Rheumatology (Oxford) 2007;46:
MG, et al. Immunosuppressive therapy in lupus nephritis: the Euro-Lupus 1096-101.
Nephritis Trial, a randomized trial of low-dose versus high-dose intravenous [252] Grootscholten C, Ligtenberg G, Hagen EC, van den Wall Bake AW, de Glas-Vos
cyclophosphamide. Arthritis Rheum 2002;46:2121-31. JW, Bijl M, et al. Azathioprine/methylprednisolone versus cyclophosphamide in
[226] Mok CC, Ying KY, Ng WL, Lee KW, CH To, Lau CS, et al. Long-term outcome of proliferative lupus nephritis. A randomized controlled trial. Kidney Int
diffuse proliferative lupus glomerulonephritis treated with cyclophosphamide. 2006;70:732-42.
Am J Med 2006;119(355):e25-33. [253] Grootscholten C, Bajema IM, Florquin S, Steenbergen EJ, Peutz-Kootstra CJ,
[227] Ioannidis JP, Katsifis GE, Tzioufas AG, Moutsopoulos HM. Predictors of sustained Goldschmeding R, et al. Treatment with cyclophosphamide delays the progres-
amenorrhea from pulsed intravenous cyclophosphamide in premenopausal sion of chronic lesions more effectively than does treatment with azathioprine
women with systemic lupus erythematosus. J Rheumatol 2002;29:2129-35. plus methylprednisolone in patients with proliferative lupus nephritis. Arthritis
[228] Boumpas DT, Austin III HA, Vaughan EM, Yarboro CH, Klippel JH, Balow JE. Risk Rheum 2007;56:924-37.
for sustained amenorrhea in patients with systemic lupus erythematosus receiv- [254] Arends S, Grootscholten C, Derksen RH, Berger SP, de Sevaux RG, Voskuyl AE,
ing intermittent pulse cyclophosphamide therapy. Ann Intern Med 1993;119: et al. Long-term follow-up of a randomised controlled trial of azathioprine/
366-9. methylprednisolone versus cyclophosphamide in patients with proliferative
[229] Mok CC, Lau CS, Wong RW. Risk factors for ovarian failure in patients with sys- lupus nephritis. Ann Rheum Dis 2011.
temic lupus erythematosus receiving cyclophosphamide therapy. Arthritis [255] Grootscholten C, Snoek FJ, Bijl M, van Houwelingen HC, Derksen RH, Berden JH.
Rheum 1998;41:831-7. Health-related quality of life and treatment burden in patients with proliferative
[230] Somers EC, Marder W, Christman GM, Ognenovski V, McCune WJ. Use of a lupus nephritis treated with cyclophosphamide or azathioprine/methylprednisolone
gonadotropin-releasing hormone analog for protection against premature ovar- in a randomized controlled trial. J Rheumatol 2007;34:1699-707.
ian failure during cyclophosphamide therapy in women with severe lupus. Ar- [256] Tse KC, Tang CS, Lio WI, Lam MF, Chan TM. Quality of life comparison between
thritis Rheum 2005;52:2761-7. corticosteroid- and -mycofenolate mofetil and corticosteroid- and -oral cyclo-
[231] Blumenfeld Z, Mischari O, Schultz N, Boulman N, Balbir-Gurman A. Gonadotro- phosphamide in the treatment of severe lupus nephritis. Lupus 2006;15:371-9.
pin releasing hormone agonists may minimize cyclophosphamide associated [257] Daleboudt GM, Berger SP, Broadbent E, Kaptein AA. Health-related quality of life
gonadotoxicity in SLE and autoimmune diseases. Semin Arthritis Rheum in patients with systemic lupus erythematosus and proliferative lupus nephritis.
2011;41:346-52. Psychol Health Med 2011;16:393-404.
[232] Soares PM, Borba EF, Bonfa E, Hallak J, Correa AL, Silva CA. Gonad evaluation in [258] Houssiau FA, D'Cruz D, Sangle S, Remy P, Vasconcelos C, Petrovic R, et al. Azathio-
male systemic lupus erythematosus. Arthritis Rheum 2007;56:2352-61. prine versus mycophenolate mofetil for long-term immunosuppression in lupus
[233] Yee CS, Gordon C, Dostal C, Petera P, Dadoniene J, Griffiths B, et al. EULAR nephritis: results from the MAINTAIN Nephritis Trial. Ann Rheum Dis 2010;69:
randomised controlled trial of pulse cyclophosphamide and methylprednisolone 2083-9.
versus continuous cyclophosphamide and prednisolone followed by azathio- [259] Dooley MA, Jayne D, Ginzler EM, Isenberg D, Olsen NJ, Wofsy D, et al.
prine and prednisolone in lupus nephritis. Ann Rheum Dis 2004;63:525-9. Mycophenolate versus azathioprine as maintenance therapy for lupus nephritis.
[234] D'Cruz DP, Houssiau FA. The Euro-Lupus Nephritis Trial: the development of the N Engl J Med 2011;365:1886-95.
sequential treatment protocol. Lupus 2009;18:875-7. [260] Zahr N, Amoura Z, Debord J, Hulot JS, Saint-Marcoux F, Marquet P, et al. Pharma-
[235] Houssiau FA, Vasconcelos C, D'Cruz D, Sebastiani GD, de Ramon Garrido E, cokinetic study of mycophenolate mofetil in patients with systemic lupus
Danieli MG, et al. Early response to immunosuppressive therapy predicts good erythematosus and design of Bayesian estimator using limited sampling strate-
renal outcome in lupus nephritis: lessons from long-term followup of patients gies. Clin Pharmacokinet 2008;47:277-84.
in the Euro-Lupus Nephritis Trial. Arthritis Rheum 2004;50:3934-40. [261] Zahr N, Arnaud L, Marquet P, Haroche J, Costedoat-Chalumeau N, Hulot JS, et al.
[236] Houssiau FA, Vasconcelos C, D'Cruz D, Sebastiani GD, de Ramon Garrido E, Mycophenolic acid area under the curve correlates with disease activity in lupus
Danieli MG, et al. The 10-year follow-up data of the Euro-Lupus Nephritis Trial patients treated with mycophenolate mofetil. Arthritis Rheum 2010;62:2047-54.
comparing low-dose and high-dose intravenous cyclophosphamide. Ann [262] Roland M, Barbet C, Paintaud G, Magdelaine-Beuzelin C, Diot E, Halimi JM, et al.
Rheum Dis 2010;69:61-4. Mycophenolate mofetil in patients with systemic lupus erythematosus: a pro-
[237] Sabry A, Abo-Zenah H, Medhat T, Sheashaa H, Mahmoud K, El-Huseini A. A com- spective pharmacokinetic study. Lupus 2009;18:441-7.
parative study of two intensified pulse cyclophosphamide remission-inducing [263] Sagcal-Gironella AC, Fukuda T, Wiers K, Cox S, Nelson S, Dina B, et al. Pharmaco-
regimens for diffuse proliferative lupus nephritis: an Egyptian experience. Int kinetics and pharmacodynamics of mycophenolic acid and their relation to
Urol Nephrol 2009;41:153-61. response to therapy of childhood-onset systemic lupus erythematosus. Semin
[238] Chan TM, Li FK, Tang CS, Wong RW, Fang GX, Ji YL, et al. Efficacy of mycophenolate Arthritis Rheum 2011;40:307-13.
mofetil in patients with diffuse proliferative lupus nephritis. Hong Kong–Guangzhou [264] Mina R, von Scheven E, Ardoin SP, Eberhard BA, Punaro M, Ilowite N, et al. Con-
Nephrology Study Group. N Engl J Med 2000;343:1156-62. sensus treatment plans for induction therapy of newly diagnosed proliferative
[239] Hu W, Liu Z, Chen H, Tang Z, Wang Q, Shen K, et al. Mycophenolate mofetil vs lupus nephritis in juvenile systemic lupus erythematosus. Arthritis Care Res
cyclophosphamide therapy for patients with diffuse proliferative lupus nephritis. 2012;64:375-83.
Chin Med J (Engl) 2002;115:705-9. [265] Baskin E, Ozen S, Cakar N, Bayrakci US, Demirkaya E, Bakkaloglu A. The use of
[240] Ong LM, Hooi LS, Lim TO, Goh BL, Ahmad G, Ghazalli R, et al. Randomized con- low-dose cyclophosphamide followed by AZA/MMF treatment in childhood
trolled trial of pulse intravenous cyclophosphamide versus mycophenolate lupus nephritis. Pediatr Nephrol 2010;25:111-7.
mofetil in the induction therapy of proliferative lupus nephritis. Nephrology [266] Chen W, Tang X, Liu Q, Fu P, Liu F, Liao Y, et al. Short-term outcomes of induction
(Carlton) 2005;10:504-10. therapy with tacrolimus versus cyclophosphamide for active lupus nephritis: a
[241] Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, et al. multicenter randomized clinical trial. Am J Kidney Dis 2011;57:235-44.
Mycophenolate mofetil versus cyclophosphamide for induction treatment of [267] Moroni G, Doria A, Mosca M, Alberighi OD, Ferraccioli G, Todesco S, et al. A random-
lupus nephritis. J Am Soc Nephrol 2009;20:1103-12. ized pilot trial comparing cyclosporine and azathioprine for maintenance therapy
[242] Ginzler EM, Dooley MA, Aranow C, Kim MY, Buyon J, Merrill JT, et al. in diffuse lupus nephritis over four years. Clin J Am Soc Nephrol 2006;1:925-32.
Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis. [268] Sisó A, Ramos-Casals M, Bové A, Brito-Zerón P, Soria N, Nardi N, et al. Outcomes
N Engl J Med 2005;353:2219-28. in biopsy-proven lupus nephritis: evaluation of 190 white patients from a single
[243] Dooley MA, Hogan S, Jennette C, Falk R. Cyclophosphamide therapy for lupus center. Med (Baltimore) 2010;89:300-7.
nephritis: poor renal survival in black Americans. Glomerular Disease Collaborative [269] Zheng Z, Zhang L, Liu W, Lei Y, Xing G, Zhang J, et al. Predictors of survival in
Network. Kidney Int 1997;51:1188-95. Chinese patients with lupus nephritis. Lupus 2012.
A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194 193

[270] Alarcón GS, McGwin G, Bertoli AM, Fessler BJ, Calvo-Alén J, Bastian HM, et al. [297] Mok CC, Ying KY, Yim CW, Ng WL, Wong WS. Very long-term outcome of pure
Effect of hydroxychloroquine on the survival of patients with systemic lupus lupus membranous nephropathy treated with glucocorticoid and azathioprine.
erythematosus: data from LUMINA, a multiethnic US cohort (LUMINA L). Ann Lupus 2009;18:1091-5.
Rheum Dis 2007;66:1168-72. [298] Illei GG, Takada K, Parkin D, Austin HA, Crane M, Yarboro CH, et al. Renal flares
[271] Kasitanon N, Fine DM, Haas M, Magder LS, Petri M. Hydroxychloroquine use are common in patients with severe proliferative lupus nephritis treated with
predicts complete renal remission within 12 months among patients treated pulse immunosuppressive therapy: long-term followup of a cohort of 145
with mycophenolate mofetil therapy for membranous lupus nephritis. Lupus patients participating in randomized controlled studies. Arthritis Rheum
2006;15:366-70. 2002;46:995–1002.
[272] A randomized study of the effect of withdrawing hydroxychloroquine sulfate in [299] Mok CC, Ying KY, Tang S, Leung CY, Lee KW, Ng WL, et al. Predictors and outcome
systemic lupus erythematosus. The Canadian Hydroxychloroquine Study Group. of renal flares after successful cyclophosphamide treatment for diffuse prolifer-
N Engl J Med 1991;324:150-4. ative lupus glomerulonephritis. Arthritis Rheum 2004;50:2559-68.
[273] Tsakonas E, Joseph L, Esdaile JM, Choquette D, Senecal JL, Cividino A, et al. A long-term [300] Moroni G, Quaglini S, Gravellone L, Gallelli B, Leoni A, Messa P, et al. Membra-
study of hydroxychloroquine withdrawal on exacerbations in systemic lupus nous nephropathy in systemic lupus erythematosus: long-term outcome and
erythematosus. The Canadian Hydroxychloroquine Study Group. Lupus 1998;7:80-5. prognostic factors of 103 patients. Semin Arthritis Rheum 2012.
[274] Pons-Estel GJ, Alarcon GS, McGwin Jr G, Danila MI, Zhang J, Bastian HM, et al. [301] Ward MM. Changes in the incidence of end-stage renal disease due to lupus
Protective effect of hydroxychloroquine on renal damage in patients with nephritis, 1982–1995. Arch Intern Med 2000;160:3136-40.
lupus nephritis: LXV, data from a multiethnic US cohort. Arthritis Rheum [302] Ward MM. Changes in the incidence of endstage renal disease due to lupus
2009;61:830-9. nephritis in the United States, 1996–2004. J Rheumatol 2009;36:63-7.
[275] Radhakrishnan J, Moutzouris DA, Ginzler EM, Solomons N, Siempos II, Appel GB. [303] Costenbader KH, Desai A, Alarcon GS, Hiraki LT, Shaykevich T, Brookhart MA,
Mycophenolate mofetil and intravenous cyclophosphamide are similar as induc- et al. Trends in the incidence, demographics, and outcomes of end-stage renal
tion therapy for class V lupus nephritis. Kidney Int 2010;77:152-60. disease due to lupus nephritis in the US from 1995 to 2006. Arthritis Rheum
[276] Chan TM, Li FK, Hao WK, Chan KW, Lui SL, Tang S, et al. Treatment of membra- 2011;63:1681-8.
nous lupus nephritis with nephrotic syndrome by sequential immunosuppres- [304] Croca SC, Rodrigues T, Isenberg DA. Assessment of a lupus nephritis cohort over
sion. Lupus 1999;8:545-51. a 30-year period. Rheumatology (Oxford) 2011;50:1424-30.
[277] Chan TM, Lin AW, Tang SC, Qian JQ, Lam MF, Ho YW, et al. Prospective controlled study [305] Faurschou M, Dreyer L, Kamper AL, Starklint H, Jacobsen S. Long-term mortality
on mycophenolate mofetil and prednisolone in the treatment of membranous and renal outcome in a cohort of 100 patients with lupus nephritis. Arthritis Care
nephropathy with nephrotic syndrome. Nephrology (Carlton) 2007;12:576-81. Res 2010;62:873-80.
[278] Yap DY, Yu X, Chen XM, Lu F, Chen N, Li XW, et al. Pilot 24 month study to com- [306] Moroni G, Quaglini S, Gallelli B, Banfi G, Messa P, Ponticelli C. The long-term out-
pare mycophenolate mofetil and tacrolimus in the treatment of membranous come of 93 patients with proliferative lupus nephritis. Nephrol Dial Transplant
lupus nephritis with nephrotic syndrome. Nephrology (Carlton) 2012;17:352-7. 2007;22:2531-9.
[279] Austin III HA, Illei GG, Braun MJ, Balow JE. Randomized, controlled trial of pred- [307] Adler M, Chambers S, Edwards C, Neild G, Isenberg D. An assessment of renal
nisone, cyclophosphamide, and cyclosporine in lupus membranous nephropa- failure in an SLE cohort with special reference to ethnicity, over a 25-year period.
thy. J Am Soc Nephrol 2009;20:901-11. Rheumatology (Oxford) 2006;45:1144-7.
[280] Navarra SV, Guzman RM, Gallacher AE, Hall S, Levy RA, Jimenez RE, et al. Efficacy [308] Contreras G, Lenz O, Pardo V, Borja E, Cely C, Iqbal K, et al. Outcomes in African
and safety of belimumab in patients with active systemic lupus erythematosus: a Americans and Hispanics with lupus nephritis. Kidney Int 2006;69:1846-51.
randomised, placebo-controlled, phase 3 trial. Lancet 2011;377:721-31. [309] Gibson KL, Gipson DS, Massengill SA, Dooley MA, Primack WA, Ferris MA, et al.
[281] Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzova D, et al. A phase III, Predictors of relapse and end stage kidney disease in proliferative lupus nephritis:
randomized, placebo-controlled study of belimumab, a monoclonal antibody focus on children, adolescents, and young adults. Clin J Am Soc Nephrol 2009;4:
that inhibits B lymphocyte stimulator, in patients with systemic lupus 1962-7.
erythematosus. Arthritis Rheum 2011;63:3918-30. [310] Austin III HA, Boumpas DT, Vaughan EM, Balow JE. Predicting renal outcomes in
[282] Rovin BH, Furie R, Latinis K, Looney RJ, Fervenza FC, Sanchez-Guerrero J, et al. Ef- severe lupus nephritis: contributions of clinical and histologic data. Kidney Int
ficacy and safety of rituximab in patients with active proliferative lupus nephri- 1994;45:544-50.
tis: the Lupus Nephritis Assessment with Rituximab study. Arthritis Rheum [311] Austin III HA, Boumpas DT, Vaughan EM, Balow JE. High-risk features of lupus
2012;64:1215-26. nephritis: importance of race and clinical and histological factors in 166 patients.
[283] Pinto LF, Velasquez CJ, Prieto C, Mestra L, Forero E, Marquez JD. Rituximab in- Nephrol Dial Transplant 1995;10:1620-8.
duces a rapid and sustained remission in Colombian patients with severe and re- [312] Pereira T, Abitbol CL, Seeherunvong W, Katsoufis C, Chandar J, Freundlich M,
fractory systemic lupus erythematosus. Lupus 2011;20:1219-26. et al. Three decades of progress in treating childhood-onset lupus nephritis.
[284] Diaz-Lagares C, Croca S, Sangle S, Vital EM, Catapano F, Martinez-Berriotxoa A, Clin J Am Soc Nephrol 2011;6:2192-9.
et al. Efficacy of rituximab in 164 patients with biopsy-proven lupus nephritis: [313] Contreras G, Pardo V, Cely C, Borja E, Hurtado A, De La Cuesta C, et al. Factors asso-
pooled data from European cohorts. Autoimmun Rev 2012;11:357-64. ciated with poor outcomes in patients with lupus nephritis. Lupus 2005;14:890-5.
[285] Aringer M, Burkhardt H, Burmester G, Fischer-Betz R, Fleck M, Graninger W, [314] Barr RG, Seliger S, Appel GB, Zuniga R, D'Agati V, Salmon J, et al. Prognosis in pro-
et al. Current state of evidence on ‘off-label’ therapeutic options for systemic liferative lupus nephritis: the role of socio-economic status and race/ethnicity.
lupus erythematosus, including biological immunosuppressive agents, in Ger- Nephrol Dial Transplant 2003;18:2039-46.
many, Austria and Switzerland — a consensus report. Lupus 2012;21:386-401. [315] Petri M, Perez-Gutthann S, Longenecker JC, Hochberg M. Morbidity of systemic
[286] Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy: a national lupus erythematosus: role of race and socioeconomic status. Am J Med
estimate of frequency in systemic lupus erythematosus and other rheumatic dis- 1991;91:345-53.
eases. Arthritis Rheum 2009;60:3761-5. [316] Korbet SM, Schwartz MM, Evans J, Lewis EJ. Severe lupus nephritis: racial differ-
[287] Merrill JT, Burgos-Vargas R, Westhovens R, Chalmers A, D'Cruz D, Wallace DJ, ences in presentation and outcome. J Am Soc Nephrol 2007;18:244-54.
et al. The efficacy and safety of abatacept in patients with non-life-threatening [317] de Castro WP, Morales JV, Wagner MB, Graudenz M, Edelweiss MI, Goncalves LF.
manifestations of systemic lupus erythematosus: results of a twelve-month, Hypertension and Afro-descendant ethnicity: a bad interaction for lupus nephritis
multicenter, exploratory, phase IIb, randomized, double-blind, placebo-controlled treated with cyclophosphamide? Lupus 2007;16:724-30.
trial. Arthritis Rheum 2010;62:3077-87. [318] Cooper GS, Treadwell EL, St Clair EW, Gilkeson GS, Dooley MA. Sociodemographic
[288] Wofsy D, Hillson JL, Diamond B. Abatacept for lupus nephritis: alternative defini- associations with early disease damage in patients with systemic lupus
tions of complete response support conflicting conclusions. Arthritis Rheum 2012. erythematosus. Arthritis Rheum 2007;57:993-9.
[289] Sciascia S, Ceberio L, Garcia-Fernandez C, Roccatello D, Karim Y, Cuadrado MJ. [319] Chen YE, Korbet SM, Katz RS, Schwartz MM, Lewis EJ. Value of a complete or partial
Systemic lupus erythematosus and infections: clinical importance of conven- remission in severe lupus nephritis. Clin J Am Soc Nephrol 2008;3:46-53.
tional and upcoming biomarkers. Autoimmun Rev 2012. [320] Moroni G, Quaglini S, Maccario M, Banfi G, Ponticelli C. “Nephritic flares” are
[290] Op De Beeck K, Vermeersch P, Verschueren P, Westhovens R, Marien G, predictors of bad long-term renal outcome in lupus nephritis. Kidney Int
Blockmans D, et al. Antinuclear antibody detection by automated multiplex im- 1996;50:2047-53.
munoassay in untreated patients at the time of diagnosis. Autoimmun Rev 2012. [321] Singh S, Zhou XJ, Ahn C, Saxena R. A retrospective analysis of clinical presenta-
[291] Reyes-Thomas J, Blanco I, Putterman C. Urinary biomarkers in lupus nephritis. tion of lupus nephritis. Am J Med Sci 2011;342:467-73.
Clin Rev Allergy Immunol 2011;40:138-50. [322] Franco C, Yoo W, Franco D, Xu Z. Predictors of end stage renal disease in African
[292] Rovin BH, Birmingham DJ, Nagaraja HN, Yu CY, Hebert LA. Biomarker discovery Americans with lupus nephritis. Bull NYU Hosp Jt Dis 2010;68:251-6.
in human SLE nephritis. Bull NYU Hosp Jt Dis 2007;65:187-93. [323] Dhir V, Aggarwal A, Lawrence A, Agarwal V, Misra R. Long-term outcome of
[293] Suzuki M, Wiers K, Brooks EB, Greis KD, Haines K, Klein-Gitelman MS, et al. lupus nephritis in Asian Indians. Arthritis Care Res 2012;64:713-20.
Initial validation of a novel protein biomarker panel for active pediatric lupus [324] Al Arfaj AS, Khalil N, Al Saleh S. Lupus nephritis among 624 cases of systemic
nephritis. Pediatr Res 2009;65:530-6. lupus erythematosus in Riyadh, Saudi Arabia. Rheumatol Int 2009;29:1057-67.
[294] Zhang X, Jin M, Wu H, Nadasdy T, Nadasdy G, Harris N, et al. Biomarkers of lupus [325] Huong DLsbLTH D, Papo T, Beaufils H, Wechsler B, Bletry O, Baumelou A, et al.
nephritis determined by serial urine proteomics. Kidney Int 2008;74:799-807. Renal involvement in systemic lupus erythematosus. A study of 180 patients
[295] Bertsias G, Ioannidis JP, Boletis J, Bombardieri S, Cervera R, Dostal C, et al. EULAR from a single center. Med (Baltimore) 1999;78:148-66.
recommendations for the management of systemic lupus erythematosus. Report [326] Patel SB, Korbet SM, Lewis EJ. The prognosis of severe lupus nephritis based on
of a Task Force of the EULAR Standing Committee for International Clinical Studies the Modification of Diet in Renal Disease (MDRD) study estimated glomerular
Including Therapeutics. Ann Rheum Dis 2008;67:195-205. filtration rate. Lupus 2011;20:256-64.
[296] Sun HO, Hu WX, Xie HL, Zhang HT, Chen HP, Zeng CH, et al. Long-term outcome [327] Faurschou M, Starklint H, Halberg P, Jacobsen S. Prognostic factors in lupus
of Chinese patients with membranous lupus nephropathy. Lupus 2008;17: nephritis: diagnostic and therapeutic delay increases the risk of terminal renal
56-61. failure. J Rheumatol 2006;33:1563-9.
194 A.T. Borchers et al. / Autoimmunity Reviews 12 (2012) 174–194

[328] Fiehn C, Hajjar Y, Mueller K, Waldherr R, Ho AD, Andrassy K. Improved clinical [348] Cauwe B, Martens E, Sagaert X, Dillen C, Geurts N, Li S, et al. Deficiency of gelatinase
outcome of lupus nephritis during the past decade: importance of early diagnosis B/MMP-9 aggravates lpr-induced lymphoproliferation and lupus-like systemic
and treatment. Ann Rheum Dis 2003;62:435-9. autoimmune disease. J Autoimmun 2011;36:239-52.
[329] Esdaile JM, Joseph L, MacKenzie T, Kashgarian M, Hayslett JP. The benefit of early [349] Lemoine S, Morva A, Youinou P, Jamin C. Human T cells induce their own
treatment with immunosuppressive agents in lupus nephritis. J Rheumatol regulation through activation of B cells. J Autoimmun 2011;36:228-38.
1994;21:2046-51. [350] Davies RC, Pettijohn K, Fike F, Wang J, Nahas SA, Tunuguntla R, et al. Defective
[330] Hsu CY, Chiu WC, Yang TS, Chen CJ, Chen YC, Lai HM, et al. Age- and gender-related DNA double-strand break repair in pediatric systemic lupus erythematosus.
long-term renal outcome in patients with lupus nephritis. Lupus 2011;20:1135-41. Arthritis Rheum 2012;64:568-78.
[331] Vachvanichsanong P, Dissaneewate P, McNeil E. Diffuse proliferative glomerulo- [351] Zhao M, Sun Y, Gao F, Wu X, Tang J, Yin H, et al. Epigenetics and SLE: RFX1
nephritis does not determine the worst outcome in childhood-onset lupus downregulation causes CD11a and CD70 overexpression by altering epigenetic
nephritis: a 23-year experience in a single centre. Nephrol Dial Transplant modifications in lupus CD4+ T cells. J Autoimmun 2010;35:58-69.
2009;24:2729-34. [352] Brooks WH, Le Dantec C, Pers JO, Youinou P, Renaudineau Y. Epigenetics and
[332] Naveed A, Nilubol C, Melancon JK, Girlanda R, Johnson L, Javaid B. Preemptive autoimmunity. J Autoimmun 2010;34:J207-19.
kidney transplantation in systemic lupus erythematosus. Transplant Proc [353] Vas J, Gronwall C, Marshak-Rothstein A, Silverman GJ. Natural antibody to
2011;43:3713-4. apoptotic cell membranes inhibits the proinflammatory properties of lupus
[333] Tang H, Chelamcharla M, Baird BC, Shihab FS, Koford JK, Goldfarb-Rumyantzev autoantibody immune complexes. Arthritis Rheum 2012.
AS. Factors affecting kidney-transplant outcome in recipients with lupus nephri- [354] Michaelson JS, Wisniacki N, Burkly LC, Putterman C. Role of TWEAK in lupus
tis. Clin Transplant 2008;22:263-72. nephritis: a bench-to-bedside review. J Autoimmun 2012.
[334] Bartosh SM, Fine RN, Sullivan EK. Outcome after transplantation of young [355] Sreih A, Ezzeddine R, Leng L, LaChance A, Yu G, Mizue Y, et al. Dual effect of the
patients with systemic lupus erythematosus: a report of the North American macrophage migration inhibitory factor gene on the development and severity
pediatric renal transplant cooperative study. Transplantation 2001;72:973-8. of human systemic lupus erythematosus. Arthritis Rheum 2011;63:3942-51.
[335] Devlin A, Waikar SS, Solomon DH, Lu B, Shaykevich T, Alarcon GS, et al. Variation [356] Zhang Q, Long H, Liao J, Zhao M, Liang G, Wu X, et al. Inhibited expression of
in initial kidney replacement therapy for end-stage renal disease due to lupus hematopoietic progenitor kinase 1 associated with loss of jumonji domain
nephritis in the United States. Arthritis Care Res 2011;63:1642-53. containing 3 promoter binding contributes to autoimmunity in systemic lupus
[336] Ishimori ML, Gudsoorkar V, Venuturupalli SR, Weisman MH. Disparities in renal erythematosus. J Autoimmun 2011;37:180-9.
replacement in lupus nephritis: current practice and future implications. Arthritis [357] Chang C. Neonatal autoimmune diseases: a critical review. J Autoimmun 2012;38:
Care Res 2011;63:1639-41. J223-38.
[337] Sule S, Fivush B, Neu A, Furth S. Increased risk of death in pediatric and adult [358] Joo H, Coquery C, Xue Y, Gayet I, Dillon SR, Punaro M, et al. Serum from patients
patients with ESRD secondary to lupus. Pediatr Nephrol 2011;26:93-8. with SLE instructs monocytes to promote IgG and IgA plasmablast differentiation.
[338] Hiraki LT, Lu B, Alexander SR, Shaykevich T, Alarcon GS, Solomon DH, et al. J Exp Med 2012;209:1335-48.
End-stage renal disease due to lupus nephritis among children in the US, [359] Biswas PS, Gupta S, Stirzaker RA, Kumar V, Jessberger R, Lu TT, et al. Dual regulation
1995–2006. Arthritis Rheum 2011;63:1988-97. of IRF4 function in T and B cells is required for the coordination of T–B cell
[339] Ward MM. Outcomes of renal transplantation among patients with end-stage interactions and the prevention of autoimmunity. J Exp Med 2012;209:581-96.
renal disease caused by lupus nephritis. Kidney Int 2000;57:2136-43. [360] Griffin DO, Rothstein TL. A small CD11b(+) human B1 cell subpopulation
[340] Bunnapradist S, Chung P, Peng A, Hong A, Lee B, Fukami S, et al. Outcomes of stimulates T cells and is expanded in lupus. J Exp Med 2011;208:2591-8.
renal transplantation for recipients with lupus nephritis: analysis of the Organ [361] Kim SJ, Zou YR, Goldstein J, Reizis B, Diamond B. Tolerogenic function of Blimp-1
Procurement and Transplantation Network database. Transplantation 2006;82: in dendritic cells. J Exp Med 2011;208:2193-9.
612-8. [362] Eilat D, Wabl M. B cell tolerance and positive selection in lupus. J Immunol
[341] Contreras G, Mattiazzi A, Schultz DR, Guerra G, Ladino M, Ortega LM, et al. Kidney 2012;189:503-9.
transplantation outcomes in African-, Hispanic- and Caucasian-Americans with [363] Chang C, Gershwin ME. Drugs and autoimmunity—a contemporary review and
lupus. Lupus 2012;21:3–12. mechanistic approach. J Autoimmun 2010;34:J266-75.
[342] Burgos PI, Perkins EL, Pons-Estel GJ, Kendrick SA, Liu JM, Kendrick WT, et al. Risk [364] Chen M, Daha MR, Kallenberg CG. The complement system in systemic autoimmune
factors and impact of recurrent lupus nephritis in patients with systemic lupus disease. J Autoimmun 2010;34:J276-86.
erythematosus undergoing renal transplantation: data from a single US institution. [365] Moreth K, Brodbeck R, Babelova A, Gretz N, Spieker T, Zeng-Brouwers J, et al. The
Arthritis Rheum 2009;60:2757-66. proteoglycan biglycan regulates expression of the B cell chemoattractant
[343] Yu TM, Wen MC, Li CY, Cheng CH, Wu MJ, Chen CH, et al. Impact of recurrent lupus CXCL13 and aggravates murine lupus nephritis. J Clin Invest 2010;120:4251-72.
nephritis on lupus kidney transplantation: a 20-year single center experience. Clin [366] Cancro MP, D'Cruz DP, Khamashta MA. The role of B lymphocyte stimulator
Rheumatol 2012;31:705-10. (BLyS) in systemic lupus erythematosus. J Clin Invest 2009;119:1066-73.
[344] Yap DY, Tang CS, Ma MK, Lam MF, Chan TM. Survival analysis and causes of [367] Berggren O, Hagberg N, Weber G, Alm GV, Ronnblom L, Eloranta ML. B lympho-
mortality in patients with lupus nephritis. Nephrol Dial Transplant 2012. cytes enhance the interferon-alpha production by plasmacytoid dendritic cells.
[345] Bono L, Cameron JS, Hicks JA. The very long-term prognosis and complications of Arthritis Rheum 2012.
lupus nephritis and its treatment. QJM 1999;92:211-8. [368] Mok CC, Wong RW, Lau CS. Lupus nephritis in Southern Chinese patients:
[346] Bogdanovic R, Nikolic V, Pasic S, Dimitrijevic J, Lipkovska-Markovic J, Eric-Marinkovic J, clinicopathologic findings and long-term outcome. Am J Kidney Dis 1999;34:
et al. Lupus nephritis in childhood: a review of 53 patients followed at a single center. 315-23.
Pediatr Nephrol 2004;19:36-44. [369] Yang LY, Chen WP, Lin CY. Lupus nephritis in children—a review of 167 patients.
[347] Al Durahim H, Al Ghamdi G, Al Seraya A, Alkhiari R, Al Sayyari A. Predictors of Pediatrics 1994;94:335-40.
mortality and end stage renal disease in Saudi patients with lupus nephritis.
Lupus 2011;20:1329-35.

S-ar putea să vă placă și