Sunteți pe pagina 1din 13

REVIEWS

Redefining lupus nephritis: clinical


implications of pathophysiologic
subtypes
Feng Yu1,2, Mark Haas3, Richard Glassock4 and Ming-Hui Zhao1,5
Abstract | Systemic lupus erythematosus (SLE) is associated with a broad spectrum of clinical and
immunologic manifestations, of which lupus nephritis is the most common cause of morbidity
and mortality. The development of nephritis in patients with SLE involves multiple pathogenic
pathways including aberrant apoptosis, autoantibody production, immune complex deposition
and complement activation. The 2003 International Society of Nephrology/Renal Pathology
Society (ISN/RPS) classification system for lupus nephritis was widely accepted with high
intraobserver and interobserver concordance to guide therapeutic strategy and provide
prognostic information. However, this classification system is not based on the underlying disease
pathophysiology. Some additional lesions that contribute to disease presentation, including
glomerular crescents, podocyte injury, tubulointerstitial lesions and vascular injury, should be
recognized. Although outcomes for patients with lupus nephritis have improved over the past
30 years, treatment of this disease remains challenging and is best approached on the basis of
the underlying pathogenesis, which is only partially represented by the various pathological
phenotypes defined by the ISN/RPS classification. Here, we discuss the heterogeneous
1
Renal Division, Department of
Medicine, Peking University mechanisms involved in the pathogenesis of lupus nephritis and how improved understanding
First Hospital, Institute of of underlying disease mechanisms might help guide therapeutic strategies.
Nephrology, Peking University,
Key Laboratory of Renal
Disease, Ministry of Health Systemic lupus erythematosus (SLE) is a chronic auto- on the 2003 International Society of Nephrology/
of China, Beijing 100034, immune disease that affects multiple organs and tis- Renal Pathology Society (ISN/RPS) classification
P. R. China. sues, of which the development of kidney disease is system1–4 (BOX 1).
2
Department of Nephrology,
Peking University International
the most important predictor of morbidity and mor- Despite the availability of these guidelines, remission
Hospital, 1 Zhongguancun Life tality. Renal involvement is indicated by the presence is achieved in only 50–70% of patients and 10–20% of
and Science Street, Changping of haematuria, proteinuria or decreased renal func- patients will progress to ESRD within 5 years of diag-
District, Beijing 102206, tion, and requires confirmation by renal biopsy. The nosis1–3. These poor outcomes represent an enormous
P. R. China.
purpose of renal biopsy is to confirm the diagnosis of challenge to the use of therapeutic strategies based on
3
Department of Pathology and
Laboratory Medicine, Cedars lupus nephritis, establish pathologic patterns, activity the current classification system5–9. Moreover, several
Sinai Medical Center, Los and chronicity of renal injury, guide therapeutic strat- clinical trials of immunomodulatory therapies, includ-
Angeles, 90048 California, egy, and provide prognostic information including the ing trials of mycophenolate mofetil10, ciclosporin11, and
USA. likelihood of response to treatment and of progression rituximab12, failed to demonstrate superiority over con-
4
Department of Medicine,
David Geffen School of
to end-stage renal disease (ESRD). A well-established ventional therapies. Although the failure of these trials
Medicine at UCLA, 8 Bethany, histopathologic classification system that is predic- might be in part attributable to the selection of incorrect
Laguna Niguel, 92677 tive of therapeutic response and outcomes is therefore end points13, their failure might be also partly attributed
California, USA. required. Three major guidelines for the management to the molecular heterogeneity and complex nature of
5
Peking-Tsinghua Center for
of lupus nephritis exist — the American College of the disease, suggesting that targeted approaches might be
Life Sciences, 5 Summer
Palace Street, Haidian District, Rheumatology (ACR) guideline, the Kidney Disease: needed to improve therapeutic efficiency in lupus nephri-
Beijing 100871, P. R. China. Improving Global Outcomes (KDIGO) guideline, tis. In support of this notion, a 2016 study that profiled
Correspondence to M.-H.Z. and the Joint European League Against Rheumatism/ the blood transcriptome of 158 paediatric patients with
mhzhao@bjmu.edu.cn European Renal Association–European Dialysis SLE confirmed a common type I interferon signature
doi:10.1038/nrneph.2017.85 and Transplant Association (EULAR/ERA–EDTA) and identified a plasmablast signature as the most robust
Published online 3 Jul 2017 ­guideline — all of which base their recommendations biomarkers of disease activity. Importantly, gradual

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 483


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points nephritis and acute kidney injury (AKI), which might
influence clinical decision making 1–3. For example, a
• The main purposes of renal biopsy in patients with systemic lupus erythematosus are 2011 analysis of a large cohort of patients with lupus
to confirm the diagnosis of lupus nephritis, to assess disease activity and/or chronicity, nephritis found evidence of pathological changes — such
guide therapeutic strategy, and provide prognostic information as endocapillary proliferative glomerulonephritis, extra-
• Although the 2003 International Society of Nephrology/Renal Pathology Society capillary proliferative glomerulonephritis (also known as
(ISN/RPS) lupus nephritis classification system has been widely accepted, some crescentic glomerulonephritis), membranoproliferative
additional lesions reflect the underlying disease pathogenesis and should be included
glomerulonephritis, thrombotic microangiopathy and
or otherwise recognized
acute tubular–interstitial nephritis — in renal biopsy
• The ISN/RPS classification system is based on histology and not necessarily on the
samples from patients with AKI21. More importantly,
underlying pathogenesis, which needs to be the focus of treatment
the different renal histopathologic injuries associated
• The presence of glomerular crescents, podocyte injury, tubulointerstitial lesions
closely with clinical implications and long-term out-
and thrombotic microangiopathy, in particular, are indicative of underlying disease
processes and should be considered when assessing patients with lupus nephritis
comes beyond serum creatinine level, highlighting the
importance of renal biopsy.
• The current management of lupus nephritis is based on steroids and non-selective
immunosuppressive drugs; novel agents that specifically interfere with the underlying
The above clinical studies illustrate the value of renal
pathophysiologic mechanisms of lupus nephritis are needed to improve disease biopsy in providing insights into disease processes that
outcomes are not apparent by clinical or laboratory data alone.
A valuable histopathologic classification system for
lupus nephritis should have certain features: it should
enrichment of neutrophil transcripts was detected dur- have clear and precise definitions of disease pathology,
ing the development of active nephritis and distinct sig- have reproducible criteria with low intraobserver and
natures in response to treatment were identified based interobserver variation, and should provide prognostic
on different pathological subtypes, enabling stratification information for response to therapy and renal outcomes.
of the patients into seven groups according to disease
activity and the underlying molecular signature14. This The 2003 ISN/RPS classification
study indicates that the pathologic classification of lupus The aim of the 2003 ISN/RPS classification of lupus
nephritis could be improved by incorporating immuno­ nephritis 4 was to accommodate new insights into
pathological and/or molecular markers of disease pro- pathologic lesions and pathogenesis of lupus nephritis
cesses with the currently used histological parameters15,16, and eliminate inconsistencies and ambiguities present in
a suggestion that is supported by other researchers17,18. the 1995 WHO classification22. Updates included a new
A good association between clinical indices and patho- definition for class I lupus nephritis, an emphasis on the
logical features is needed to evaluate the treatment diagnosis of combinations of membranous and prolif-
response of patients with lupus nephritis19. erative glomerulonephritis (class III and V, or class IV
In this Review, we summarize potential approaches and V), the separation of class IV lupus nephritis into
by which the ISN/RPS classification system could be segmental (IV‑S) and global (IV‑G) variants, and clearer
improved through consideration of underlying disease definitions for all classes4 (BOX 1).
processes characterized by the presence of glomerular Since its publication multiple validation studies have
crescents, podocyte injury, tubulointerstitial lesions and compared the ISN/RPS classification with the older WHO
vascular injury. Particular emphasis is given to discus- classification. One comparative study reported higher
sion of the potential clinical relevance of histopathologic reproducibility using the ISN/RPS classification, but
findings in predicting disease outcome and in facilitating noted that the interobserver agreement was far from ideal
the choice of therapeutic interventions. and that reproducibility of the assessment of activity and
chronicity scores was disappointing 23. Similarly, a 2008
Classification of lupus nephritis study reported that intraobserver and inter­observer
The value of renal biopsy agreement was higher using the ISN/RPS classification
Renal biopsy is crucial to the diagnosis and management than the WHO classification, although interobserver
of lupus nephritis. The KDIGO, ACR and EULAR/ERA– agreement was found to be low for both classification sys-
EDTA guidelines recommend that a renal biopsy is per- tems24. Studies have also evaluated the prognostic value
formed to confirm the diagnosis, assess disease activity of the 2003 classification system; for instance, one retro-
and/or chronicity and guide treatment 1–3. A repeat renal spective study of long-term outcomes of 60 patients with
biopsy should be considered in some circumstances such lupus nephritis reported good correlation between disease
as in patients who relapse, or to reconcile discordance classification according to the ISN/RPS system and long-
among clinical parameters such as persisting immuno- term renal outcomes in terms of predicting response to
logical activity despite complete or partial remission of therapy, and progression to ESRD and/or death25. Many
proteinuria or renal function3,4,20. In general, renal biopsy other studies since 2005 have further documented the
should be considered when a patient with SLE develops importance of renal biopsy and the ISN/RPS system in
proteinuria (>500 mg per day or >3+ on urine dipstick), managing patients with lupus nephritis19,26–32.
active urine sediments or evidence of renal insufficiency Although the ISN/RPS classification is an improve-
(defined by an estimated glomerular filtration rate ment on previous systems and has been widely accepted
<60 ml/min/1.73 m2). Some clinical analyses have also by renal pathologists and clinicians, it relies heavily on
demonstrated histological variants in patients with lupus pathology as observed by light microscopy, and not the

484 | AUGUST 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | The 2003 ISN/RPS lupus nephritis classification system4 C4d, C3 and the membrane attack complex, in the classi-
fication17,18, given evidence for the involvement of com-
Class I: Minimal mesangial lupus nephritis plement in disease pathogenesis. Here, we outline some
• Normal glomeruli by light microscopy, but mesangial immune deposits by of the key issues with the current classification system
immunofluorescence. and suggest additional lesions that should be considered
Class II: Mesangial proliferative lupus nephritis in any new classification system of lupus nephritis.
• Purely mesangial hypercellularity of any degree or mesangial matrix expansion by
light microscopy, with mesangial immune deposits. Cut-off thresholds for segmental versus global d ­ iffuse
• A few isolated subepithelial or subendothelial deposits may be visible by nephritis. The ISN/RPS classification divides class IV
immunofluorescence or electron microscopy, but not by light microscopy. lupus nephritis into IV‑S and IV‑G subclasses, depend-
ing on whether the glomerular lesions are primarily
Class III: Focal lupus nephritis*
segmental or global4. This subclassification of class IV
• Active or inactive focal, segmental or global endocapillary or extracapillary
lupus nephritis was initially proposed on the basis of
glomerulonephritis involving <50% of all glomeruli, typically with focal
subendothelial immune deposits, with or without mesangial alterations. findings showing that diffuse segmental lesions had
more vasculitic–like features than global lesions and
Class IV: Diffuse lupus nephritis‡ were associated with a worse renal prognosis42. However,
• Active or inactive diffuse, segmental or global endocapillary or extracapillary subsequent validation studies that have compared fea-
glomerulonephritis involving ≥50% of all glomeruli, typically with diffuse tures of IV‑S with IV‑G have failed to show statistically
subendothelial immune deposits, with or without mesangial alterations.
significant differences in renal outcomes between the
• This class is divided into diffuse segmental (IV‑S) lupus nephritis when ≥50% of the two subclasses24,25,43–46, a finding supported by a 2012
involved glomeruli have segmental lesions, and diffuse global (IV‑G) lupus nephritis
­meta-analysis of eight studies47.
when ≥50% of the involved glomeruli have global lesions. Segmental is defined as a
glomerular lesion that involves less than half of the glomerular tuft. This class includes
The subclassification of ISN/RPS class IV nephritis,
cases with diffuse wire loop deposits but with little or no glomerular proliferation. which includes all biopsy samples with ≥50% glomeru-
lar involvement, into IV‑S and IV‑G lesions differs from
Class V: Membranous lupus nephritis the original 1982 WHO classification of lupus nephritis,
• Global or segmental subepithelial immune deposits or their morphologic sequelae by which classified biopsy samples according to the type of
light microscopy and by immunofluorescence or electron microscopy, with or without lesion: those with severe segmental glomerulonephri-
mesangial alterations.
tis with ≥50% glomerular involvement were classed as
• Class V lupus nephritis may occur in combination with class III or IV in which case both WHO class III ≥50%, and those with diffuse global glo-
will be diagnosed.
merulonephritis were classed as WHO class IV48. A 2008
• Class V lupus nephritis may show advanced sclerosis. study in which biopsy samples with WHO class IV or
Class VI: Advanced sclerotic lupus nephritis class III >50% lupus nephritis were reclassified accord-
• ≥90% of glomeruli globally sclerosed without residual activity. ing to ISN/RPS criteria led to the reclassification of 39
Indicate and grade (mild, moderate, severe) tubular atrophy, interstitial inflammation and
WHO class IV samples as ISN/RPS class IV-G46. Of the
fibrosis, severity of arteriosclerosis or other vascular lesions. *Indicate the proportion of 44 samples originally classified as WHO class III >50%,
glomeruli with active and with sclerotic lesions. ‡Indicate the proportion of glomeruli with 22 were reclassified as ISN/RPS IV‑S. The remaining 22
active and with sclerotic lesions, and the proportion of glomeruli with fibrinoid necrosis and/or were considered to be ISN/RPS class IV‑G lupus nephri-
cellular crescents. Modified with permission from the International Society of Nephrology ©
tis, but were categorized separately as class IV‑query
Weening, J. J. et al. Kidney Int. 65, 521–530 (2004).
(IV‑Q). Of the 61 patients with class IV‑G, renal sur-
vival was worse for those with class IV‑Q (that is, those
underlying disease pathophysiology. Although small who had been reclassified from the WHO class III >50%
studies are available, whether the classification system category) than for those originally classified as WHO
can accurately predict clinical outcomes and thera- class IV (33% versus 77% at 10 years, P = 0.001). 10‑year
peutic responses when combined with clinical and renal survival was almost twice as high for patients in
laboratory data has not been thoroughly assessed17. class IV‑S than for those with class IV‑Q, but did not quite
Prospective, evidence-based validation studies similar reach statistical significance (62% versus 33%, P = 0.057).
to those used to validate the Oxford MEST system for The researchers concluded that although the ISN/RPS
IgA ­nephropathy 33–41 are still needed. IV‑S and IV‑G categories were created to be analogous
to severe segmental and diffuse glomerulonephritis in
Proposed modifications the WHO classification and are considered to be equiva-
In 2015, a group of nephropathologists highlighted lent by some pathologists, clearly they are not. Grouping
some difficulties with the ISN/RPS classification, mostly patients who were originally classified as having WHO
relating to uncertainties in the evaluation of particular class III >50% (that is, those with IV‑Q) together with
disease classes and inconsistencies in the definitions of those with the best prognosis (WHO class IV) within
Segmental lesions histologic parameters16. The researchers also suggested subclass IV‑G, obscures prognostic and pathogenic dif-
A lesion that involves less than ways in which the classification could be improved, in ferences, and explains the lack of difference in outcomes
half the glomerular tuft. particular, by including thrombotic microangiopathy between ISN/RPS IV‑S and IV‑G lesions in the studies
and glomerular crescents in the classification system mentioned above.
Global lesions
A glomerular lesion that affects
because these lesions might adversely affect renal out- The threshold of 50% glomerular involvement to
more than 50% of the comes16. Other researchers have suggested incorporat- define classes III and IV and the use of segmental and
glomerular tuft. ing components of the complement system, such as C1q, global lesions to subdivide class IV lupus nephritis in the

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 485


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

ISN/RPS system were predominantly selected to bring biopsy samples, suggesting further similarities between
the classification in line with traditional pathology ter- crescentic IV‑G samples and ANCA vasculitis. These
minology, rather than based on actual data relating to findings suggest that differences in underlying patho-
the clinical relevance of these features; however, some genic mechanisms and disease outcomes might exist
studies published since publication of the 2003 ISN/RPS within the same subclass of lupus nephritis, and leads
classification support the notion that pathological dif- us to recommend that the presence of true crescentic
ferences exist between IV‑G and IV‑S lesions. A 2005 glomerulonephritis (defined as the presence of crescents
study demonstrated that class IV‑S lesions had more glo- in ≥50% of glomeruli) is included as part of a new lupus
merular fibrinoid necrosis and fewer immune deposits nephritis classification system (FIG. 1).
than IV‑G lesions, whereas IV‑G lesions behaved as a
typical immune complex-mediated glomerulonephri- Podocyte injury. Renal biopsy samples from patients
tis, suggesting pathogenic differences44. A study of auto­ with lupus nephritis can demonstrate injury to nearly
antibodies supports this hypothesis. We demonstrated any cell type, including the podocyte. Evidence for the
a higher prevalence of anti-neutrophil cytoplasmic clinical relevance of podocyte injury comes from several
antibodies (ANCA) in sera of patients with class IV‑S studies that have described patients with lupus nephritis,
lupus nephritis than in those with class IV‑G nephri- nephrotic-range proteinuria and extensive effacement of
tis and a higher prevalence of anti‑C1q IgG1 and IgG3 podocyte foot processes, without evidence of immune
subclasses in sera of patients with class IV‑G nephri- complex deposition in peripheral glomerular capillar-
tis45. Together, these data suggest that some forms of ies or endocapillary proliferation52,53. Morphologically,
lupus nephritis might resemble ANCA-associated these podocyte lesions resemble minimal change dis-
­ auci-­i mmune necrotizing and crescentic glomeru-
p ease or focal segmental glomerulosclerosis (FSGS), and
lonephritis (as is discussed in further detail below). researchers involved in these studies have proposed
However, ­studies27,43,44 have also shown that class IV‑S using the term ‘lupus podocytopathy’ to describe this
and IV‑G lupus nephritis can transform between each entity 54–56. A retrospective study of 19 patients with col-
other, potentially challenging the concept that different lapsing glomerulopathy and SLE or SLE-like disease
pathogenic mechanisms underlie the two subclasses. reported massive proteinuria in 95% of patients; seg-
Re‑evaluation of the ISN/RPS classification should con- mental and/or global collapsing glomerulopathy was
sider a more evidence-based approach to define cut-off seen in 11–77% of glomeruli, and extensive foot pro-
thresholds for focal versus diffuse lesions as well as the cess effacement was seen in 82% of patients. Seven of
pathophysiological and clinical relevance of segmental 13 patients with follow‑up data progressed to ESRD57.
versus global forms of lupus nephritis. A large Chinese study of 3,750 biopsy samples from
patients with lupus nephritis identified 50 cases (1.33%)
Crescentic lupus nephritis in class IV‑G. Rapidly pro- of lupus podocytopathy, including 13 cases of minimal
gressive glomerulonephritis (RPGN) is characterized change disease, 28 instances with evidence of mesangial
by pathological features of crescentic glomerulonephri- proliferation, and nine cases of FSGS58. Extensive foot
tis and is a severe clinical syndrome associated with process effacement was evident in all 50 biopsy sam-
progressive loss of renal function. Crescents can be ples and electron-dense deposits present exclusively in
observed in association with a number of different renal mesangial cells were evident in 47 biopsy samples. All
pathologies, and are fairly common in biopsy samples 50 patients presented with nephrotic syndrome, and
from patients with lupus nephritis49. In one cohort of 62 34% had AKI. Most notably, patients with FSGS had a
patients with RPGN and biopsy-proven crescentic glo- higher risk of AKI and severe tubulointerstitial injury
merulonephritis, 32 patients (51.6%) had lupus nephri- and a lower remission rate than patients with minimal
tis50. The clinical relevance of these crescents is, however, change disease or mesangial proliferation, although
unclear as no studies have examined the clinical rele- no patient died or developed ESRD over a median fol-
vance of extensive (or true) crescentic lupus nephritis low‑up of 62 months. On the basis of the differences in
(which we define as the presence of crescents in ≥50% AKI incidence, severity of tubular injury and response
of glomeruli). In a cohort of 152 Chinese patients with to treatment, the researchers proposed that lupus podo­
class  IV‑G lupus nephritis, we found that patients cytopathy is divided into two different subtypes: m­ inimal
with extensive crescents (affecting ≥50% of glomeruli) change/mesangial proliferation and FSGS58.
have worse outcomes than those of other patients with A 2014 study 59 of renal biopsy samples from patients
class IV‑G lupus nephritis51. with lupus nephritis and proteinuria found that
Although biopsy samples with true crescentic glo- immuno­histochemical staining for podocyte markers
merulonephritis are classified as diffuse class IV lupus was generally preserved in patients with membranous
nephritis according to the ISN/RPS schema, in our histo- (class V) lupus nephritis. By contrast, <10% of prolifer-
pathological evaluations51, we found that biopsy samples ative forms of lupus nephritis (class III or IV) showed
with crescentic IV‑G lesions have significantly higher preserved expression of podocyte markers, suggesting
Pauci-immune levels of interstitial inflammation, interstitial fibrosis and the presence of structural podocyte damage in prolif-
A pattern associated with necrosis than IV‑G biopsy samples without crescentic erative forms of lupus nephritis. This loss of podocyte
minimal evidence of staining
for immunoglobulins on
glomerulonephritis51. More interestingly, we identi- integrity in patients with proliferative forms of lupus
glomeruli by fied lower levels of IgG, IgA, IgM, C3, C1q and fibrin nephritis might account for the worse prognosis of
immunofluorescence. in crescentic biopsy samples than in non-crescentic these patients compared to those with membranous

486 | AUGUST 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

lupus nephritis, despite similar levels of proteinuria, at in 202 Chinese patients with lupus nephritis. FPW cor-
least in the patients in this cohort. Our study measured related with proteinuria, and a threshold FPW was iden-
foot process width (FPW) to assess the podocyte foot tified that could differentiate nephrotic proteinuria from
process effacement and studied correlations between non-nephrotic proteinuria60. On the basis of these stud-
podocyte damage and clinico-pathological parameters ies, we suggest that the presence of lupus podocytopathy,

a b Podocyte injury

Crescentric
glomerulonephritis

c Mesangial hypercellularity

Podocyte

Mesangial
cell

d Diffuse lupus
Endothelial nephritis
cell

Parietal Bowman
epithelial capsule IV-G IV-S
cell

e Vascular lesions

Blood
vessel

Blood clot Plaque ICDs


Plaque
f Tubulointerstital ICDs Interstitial Tubulitis Arteriosclerosis TMA
lesions inflammation

T cell

Macrophage

Figure 1 | Pathological features of lupus nephritis subtypes. a | The hypercellularity with infiltration of inflammatory cellsReviews
Nature (PAS staining ×400).
| Nephrology
proliferation of extracapillary epithelial cells and infiltration of inflammatory e | Vascular lesions include the presence of immune complex deposits (ICDs)
cells leads to the formation of cellular crescents. Periodic acid–silver in the vasculature, demonstrated by granular staining for IgG along
methenamine staining (PASM) ×400. b | Podocyte injury and foot process arteriolar walls (immunofluorescence ×400), arteriosclerosis characterized
effacement can also occur in lupus nephritis. Original magnification of by arterial intimal fibrosis (PASM staining ×400), and thrombotic
electron microscope image ×6,000. c | Mesangial hypercellularity is defined microangiopathy (TMA) with arteriolar thrombi and swelling of endothelial
by the proliferation of mesangial cells, matrix expansion and mesangial cells (PASM staining ×400). f | Tubulointerstitial lesions include ICDs in the
deposition of immune complexes. Periodic acid–Schiff (PAS) staining ×400. tubular basement membrane demonstrated by granular staining for IgG
d | Diffuse (class IV) lupus nephritis can be subsclassified into segmental (immunofluorescence ×400), interstitial inflammation with severe interstitial
(IV‑S) and global (IV‑G) subclasses. Class IV‑S exhibits segmental fibrinoid infiltration of mononuclear inflammatory cells (haematoxylin and eosin
necrosis with segmental endocapillary hypercellularity (Masson trichrome staining ×400), and tubulitis with infiltration of lymphocytes between
staining ×400). Class IV‑G exhibits global endocapillary and mesangial tubular epithelial cells (PAS staining ×400).

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 487


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

defined by the presence of nephrotic-range protein­uria vasculopathy (3.8%), and true renal vasculitis (0.6%);
and podocyte lesions, might represent an extreme form 37.6% of the patients presented with more than two
of podocyte damage. We propose that the degree of types of vascular lesions69. Patients with TMA had the
podocyte injury in different types of lupus nephritis poorest renal outcome; surprisingly, those with purely
should be given more attention (FIG. 1), as the presence vascular ICDs (that is, with no other vascular lesion)
of specific injuries might have t­ herapeutic implications. had the second-to‑worst outcomes, although vascular
ICDs are traditionally thought to be a benign pheno-
Tubulointerstitial lesions. The ISN/RPS classification type69. A series of other studies indicate that renal vas-
focuses primarily on glomerular pathology, although it cular lesions are closely associated with clinical disease
indicates that the biopsy report should include a descrip- activity and renal outcomes, and that renal TMA is an
tion of tubulointerstitial injuries. An increasing body independent risk factor for long-term renal outcomes
of evidence, however, suggests that the importance of in patients with lupus nephritis69,71–74. The presence of
tubulo­interstitial damage requires greater emphasis than TMA lesions in renal biopsy samples from patients with
is given in the current classification. We investigated the lupus nephritis might not always be associated with
impact of tubulointerstitial damage in a multicentre haematological features of microangiopathy, such as
cohort of 313 lupus nephritis patients from North China, haemolytic anaemia, schistocytosis and thrombocyto-
and found that glomerular and tubulointerstitial lesions paenia). Thus, we propose that concise descriptions and
do not always coexist, and that tubulointerstitial inflam- grades of different renal vascular lesions are added to the
mation, tubular atrophy, and interstitial fibrosis were classification system.
independent risk factors for renal outcomes61. In another In another study of 79 patients with biopsy-proven
cohort of 68 patients from the USA, Hsieh et al. found lupus nephritis, we found that 50 (63.3%) had arterio-
that 72% of biopsy samples had evidence of moderate or sclerosis lesions on renal biopsy whereas 29 patients had
severe tubulointerstitial inflammation62. The research- no evidence of vascular changes75. Patients with arterio-
ers found that severity of tubulointerstitial inflamma- sclerosis presented with more severe echocardiographic
tion, but not severity of glomerular injury, identified indices, including larger left atrial diameter, left ven-
patients at greatest risk of ESRD. Similar results have tricular end-diastolic diameter and interventricular sep-
been reported by several other studies63–67. Hsieh et al. tum thickness, than patients without vascular changes,
also suggested that patients with lupus nephritis and highlighting the clinical relevance of vascular lesions75.
tubulointerstitial inflammation should receive inten-
sive therapeutic intervention62. To this end, a 2013 Disease pathogenesis and treatment
cohort study of 73 patients with lupus nephritis iden- Despite the use of histopathology to guide therapeutic
tified a correlation between interstitial inflammation at decisions, the morbidity and mortality of lupus nephritis
repeat biopsy and renal survival (P = 0.005), despite the remains high. The current approach to the management
absence of a correlation between interstitial inflamma- of lupus nephritis is based on steroids and other non-
tion at the baseline biopsy and worsening of renal func- specific immunosuppressive drugs. Dysregulation of
tion (P = 0.17)63. Furthermore, resolution of interstitial the immune system is fundamental to the pathogenesis
inflammation in lupus nephritis as evident on the repeat of lupus nephritis, and targeting multiple aspects of the
biopsy correlated with a favourable outcome in patients immune response through the combined use of multiple
with interstitial inflammation at baseline (P = 0.047). immunosuppressants (multitarget therapy) proved supe-
Thus, thorough assessment of tubulointerstitial dam- rior to a standard regimen of steroids and cyclophos-
age provides information about disease prognosis and phamide (intravenous or oral) as induction therapy for
should be included in the lupus nephritis classification lupus nephritis in a 2015 Chinese study 76. Nonselective
system (FIG. 1). immuno­suppressants are, however, associated with
potentially life-threatening complications including an
Renal vascular lesions. The ISN/RPS classification increased risk of infection. In addition, they are often
requests that pathologists indicate the severity of arterio­ associated with incomplete renal remission and a high
sclerosis or other vascular lesions, but overall pays very rate of renal flares, which might be more prominent in
little attention to vascular lesions, although these are patients with features such as vasculopathy, crescentic
common in biopsy samples from patients with lupus glomerulonephritis, and tubulointerstitial lesions1–3.
nephritis68. Two studies from 2012 and 2013 showed Thus, treatment of lupus nephritis remains a challenge
that adding an assessment of vascular damage to the and we propose that it should target the pathogenesis
ISN/RPS classification increased its prognostic value69,70. of the disease, as determined by the assessment of the
At least five renal vascular lesions are commonly histopathological disease phenotypes.
observed in biopsy samples from patients with lupus Novel drugs that interfere specifically with the patho-
nephritis: vascular immune complex deposits (ICDs), genic mechanisms of lupus nephritis, with fewer adverse
arteriosclerosis, thrombotic microangiopathy (TMA), effects and higher efficacy than nonspecific immuno-
non-inflammatory necrotizing vasculopathy and true suppressive agents, are needed. Here, we briefly dis-
Renal flares renal vasculitis68 (FIG. 1). In a cohort of 341 patients with cuss traditional therapeutic approaches guided by the
Defined by an increase in urine
sediment, protein excretion,
stable lupus nephritis, we found renal vascular lesions ISN/RPS classification system, and how consideration
and serum creatinine value in 81.8% of samples, including vascular ICDs (74.3%), of additional lesions might facilitate the identification of
from baseline. arteriosclerosis (24.0%), TMA (17.6%), necrotizing more targeted therapies77–83.

488 | AUGUST 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Tertiary lymphoid organs Current therapeutic approaches of these three guidelines, the general recommendations
Highly organized lymph As mentioned earlier, the three major guidelines, are outlined below along with brief discussion of the
node-like structures. ACR, KDIGO and EULAR/ERA–EDTA1–3, base their underlying pathogenesis of the different subtypes.
recommendations primarily on the underlying histo-
logic lesions as defined by the ISN/RPS classification. Class I and Class II lupus nephritis. Class I and Class II
Although some differences exist in the recommendations lupus nephritis result from immune complexes that
form within the mesangium by binding of antibodies to
Immunostimulatory autoantigens or following the release of antigens from
Dendritic cell nucleic acids mesangial cells84.
BAFF
The deposition of immune complexes in the mesan-
BAFF-R CD40 CD40L gium and expansion of the mesangial matrix do not
usually cause irreversible glomerulosclerosis, possibly
CD19 MHC class II IL-23 owing to the regenerative capacity of mesangial cells84.
B cell T cell TH17 Thus, Class I and Class II lupus nephritis might imply
TCR IL-17 a more favourable prognosis than other classes of lupus
IL-6
B7 CD28 nephritis, and aggressive immunosuppressive therapy is
CTLA4
CD20 not indicated unless the proteinuria is >3 g per day or
CD22
HMGB1 extrarenal clinical manifestations are prominent.
Plasma Macrophage histones Treg
cell Class III and IV lupus nephritis. Class III and Class IV
Immune Neutrophil lupus nephritis result from the deposition of immune
Autoantibodies complexes
complexes in the subendothelial space of the glomeru-
Autoantigen- lar capillaries, which causes endothelial cell activation
Autoantigens TLR specific T cell via the same complement Fc receptor and nucleic acid
Endothelial cell sensing mechanisms that operate in mesangial cells
GBM (FIG. 2). The development of tertiary lymphoid organs in
the tubulointerstitium leads to clonal expansion and
ongoing somatic hypermutation of B cells and plasma
cells that can lead to intrarenal autoantibody production,
Podocyte C4d
TH17 local inflammation, and tissue pathology, implying that
Complement B cell-targeted therapies with heightened tissue penetra-
TNF tion might be beneficial for patients with class III and IV
Epithelial cell IFNα lupus nephritis85,86.
The recommended therapy for class III and IV lupus
nephritis includes a sequence of induction and main­ten­
Interstitial
ance phases. Induction therapy usually involves a combi-
endothelial cell nation of high-dose parenteral and oral corticosteroids
with either cyclophosphamide (oral or intravenous) or
Antiphospholipid
mycophenolate mofetil (MMF), whereas maintenance
Tubular
Anti-dsDNA epithelial cell antibodies therapy involves azathioprine or MMF and low-dose oral
antibodies
corticosteroids. Studies have reported average remission
BCL-2 T cell (complete and partial) rates of nearly 70% after 6 months
Blood vessel initial therapy 10,87,88, but higher remission rates can be
achieved with more prolonged treatment.
Figure 2 | Cell-mediated disease mechanisms of lupus nephritis. The sensing of A 2012 systematic review found that MMF in
Nature Reviews
immunostimulatory nucleic acids by dendritic cells drives B lymphocyte and | Nephrology
combination with corticosteroids was as effective as
T lymphocyte activation and the production of autoantibodies and autoantigen-
specific T cells, leading to glomerular endothelium, podocyte, tubulointerstitial and cyclophosphamide plus corticosteroids in achieving
vascular injury. Specific leukocyte subsets, including IL‑17‑producing T helper type 17 remission in patients with proliferative lupus nephritis.
(TH17) cells, drive inflammation and contribute to renal immunopathology. B‑cell- However, these standard treatments are all associated
activating factor (BAFF) might increase the generation of new autoreactive B cells, with considerable adverse effects, therapy failure and
inhibitB cell apoptosis and stimulate the differentiation of B cells into immunoglobulin- relapse, highlighting the need for more effective, less
producing plasma cells. Infiltrating leukocytes might also provide a source of nuclear toxic options. Interest is growing in the potential use
antigens and cytokines, such as interferon (IFN) α, tumour necrosis factor (TNF), IL‑1 and of calcineurin inhibitors (CNIs) in severe class III and
IL‑6. High mobility group box 1 (HMGB1), biglycan and histones released from immune IV lupus nephritis1–3. A 2015 meta-analysis reported
cells can bind Toll-like receptors (TLRs) expressed by endothelial cells, leading to an that tacrolimus was more effective than cyclophospha-
inflammatory response. Inaddition, anti-double-stranded DNA (anti-dsDNA) antibodies
mide at inducing complete remission in patients with
can induce fibronectin secretion in proximal renal tubular epithelial cells, leading to
TGFβ activation and collagen synthesis. Activation of the apoptosis regulator BCL‑2 class III and IV lupus nephritis, but was not more effec-
might also contribute to the development of tubulointerstital inflammation, whereas tive than MMF89. However, randomized clinical trials of
antiphospholipid antibody-induced thrombosis might contribute to the development of tacrolimus in lupus nephritis have so far been small90,91.
interstitial vascular inflammation. GBM, glomerular basement membrane; TCR, T‑cell Extended follow‑­­up of the CYCLOFA-LUNE trial
receptor; Treg, regulatory T cell. (median 7.7 years) indicated that the CNI ciclosporin

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 489


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

with regard to the use of CNIs in patients with lupus


Tabalumab BAFF
nephritis, especially in those with renal vascular lesions
such as TMA, which has been attributed to the use of
Belimumab BAFF-R CD40 CD40L drugs such as CNIs. Large-scale, multicentre clinical
Nonselective immunosuppressants

CD19 MHC class II trials are needed to further confirm the safety and effi-
cacy of CNIs in lupus nephritis: the ongoing AURA‑LV
B cell T cell
TCR study 92 will hopefully provide insights into the safety and
efficacy of the new CNI, voclosporin in patients with
B7 CD28 class III and IV lupus nephritis93.
CD20 CD22 CTLA4
IL-23 Of note, and as discussed in further detail below,
lupus nephritis remission is largely defined on the basis
Rituximab Epratuzumab Abatacept Anti-IL-23 of a decline in proteinuria, and all CNIs act directly on
Plasma cell IL-6 podocytes to reduce proteinuria independent of their
Autoantibodies Immune complexes immunosuppressive effects2 (FIG. 3). Thus, whether the
Sirukumab
disease remissions observed with CNI therapy result
Autoantigens from true remission of the immunological disease pro-
Endothelial cell cesses or are the result of the antiproteinuric effects of
GBM CNIs is not clear. Repeat renal biopsies are needed to
make this distinction.
Podocyte
Class V lupus nephritis. Class V lupus nephritis (also
C4d IL-17 known as membranous lupus nephritis) is caused by
TH17
Eculizumab/ the deposition of immune complexes in the subepithe-
CNIs

Neutrophil
CCX-168 lial compartment of the glomerular tuft, which leads to
Complement
Macrophage
complement activation and ultimately podocyte injury 94
Infliximab TNF (FIG. 2). Patients with class V lupus nephritis are usually
Autoantigen- treated with antiproteinuric and antihypertensive med-
Anti-IFN-γ IFNα specific T cell ications such as renin–angiotensin system blockers,
and can receive corticosteroids and immunosuppres-
sants as required depending on the presence of persis-
Epithelial cell tent nephrotic proteinuria or extrarenal manifestations
T cell BCL-2 ABT-199
of SLE.
As noted above, CNIs can reduce proteinuria through
Figure 3 | Therapeutic targets in systemic lupus erythematosus Nature(SLE)
Reviews Nephrology
and |lupus direct actions on podocytes. CNIs inhibit dephosphoryl-
nephritis. Greater understanding of the pathogenic processes underlying lupus nephritis ation and degradation of the actin-associated podocyte
has led to the identification of new therapeutic targets. B cells have a critical role in the
protein, synaptopodin, and thus help to stabilize the
pathogenesis of SLE, and B‑cell depletion therapy therefore remains an attractive
therapeutic option. The anti‑CD20 antibody, rituximab, can deplete autoreactive
podocyte actin cytoskeleton. Preliminary data indi-
B cells and thereby attenuate the production of autoantibodies involved in disease cate that combination CNI and corticosteroid therapy
manifestations. The B‑cell-activating factor (BAFF)-neutralizing antibody, belimumab, is effective in the treatment of class V lupus nephri-
was shown to reduce renal flares and proteinuria in patients with SLE. Other promising tis, especially in reducing proteinuria89,94–97; however,
B cell-depleting monoclonal antibodies, such as tabalumab and epratuzumab, require the initiation and duration of CNI therapy remains
further testing. Abatacept is a co-stimulatory inhibitor that targets B7‑1 (CD80) on the a matter of debate, as relapses are very common once
surface of dendritic cells or B cells, and blocks co-stimulation of CD28 on T cells. CNIs are stopped2. Moreover, CNIs are associated with
Inhibitors of the complement system, such as eculizumab and CCX‑168 could have an increased risk of TMA, indicating that alternative
therapeutic value in patients with concurrent thrombotic microangiopathy or ­therapeutic approaches are required.
anti-neutrophil cytoplasmic antibody-associated vasculitis. Calcineurin inhibitors (CNIs)
have immune modulatory effects but also direct effects on podocytes, and might be
useful in patients with lupus-associated podocyte injury. Inhibition of the apoptosis
Class VI lupus nephritis. The development of sclerotic
regulator BCL‑2 prevented the development of tubulointerstital inflammation in a mouse lesions in lupus nephritis results mainly from insuffi-
model of lupus nephritis. IL‑23 activation might have a role in the development of cient cellular regeneration following the injury and loss
glomerular crescents, suggesting that anti‑IL‑23 antibodies might be beneficial. Other of epithelial cells98, and leads to irreversible glomerulo­
promising therapies that need further exploration include infliximab, which targets the sclerosis and nephron loss. Vascular rarefaction, tubulo­
inflammatory tumour necrosis factor (TNF), sirukumab, which targets inflammatory IL‑6, interstitial ischaemia, and inflammation all contribute
and monoclonal antibodies to interferon (IFN). GBM, glomerular basement membrane; to the progression of renal fibrosis and sclerosis, which
TH17, T helper type 17 cell. is usually associated with a progressive decline in glo-
merular filtration rate and ultimately the development
of ESRD.
achieved similar results to those achieved with cyclo- Patients with class VI lupus nephritis should be
phosphamide in patients with class III and IV lupus treated with renin–angiotensin system blockers.
nephritis11. Although these small trials did not find any Corticosteroids and immunosuppressive agents should
Vascular rarefaction obvious evidence of acute or chronic nephrotoxicity be used only as dictated by the presence of extrarenal
Loss of capillaries. with use of tacrolimus or ciclosporin, concerns still exist manifestations.

490 | AUGUST 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Treatments guided by additional lesions nephritis and severe podocyte effacement, we found that
As described above, the presence of features beyond those who received CNIs had better remission rates and
those included in the current classification system of long-term renal outcomes than those treated with other
lupus nephritis — including the presence of glomerular regimens, suggesting that therapies that facilitate podo-
crescents, podocyte injury, vasculopathy and tubulo­ cyte stability might be beneficial in a subgroup of patients
interstitial changes — provide insights into the under­ with lupus nephritis60 (FIGS 3,4).
lying pathogenic mechanisms of lupus nephritis and
might aid the identification of new therapeutic targets. Tubulointerstitial lesions. The mechanisms underlying
Here we discuss the underlying pathogenic mechan­ the development of tubulointerstitial lesions in lupus
isms of each of these lesions and how they might be nephritis remain unclear, and thus no specific treat-
targeted therapeutically. ment options exist. Severe tubulointerstitial inflamma-
tion is associated with in situ adaptive immunity and the
Crescentic lupus nephritis. The mechanisms that drive development of tertiary lymphoid organ–like structures
the formation of crescents in lupus nephritis are not with aggregates of T cells and B cells, plasmablast foci,
fully understood. A 2009 study showed that depletion and germinal centres110. Vimentin, an antigenic feature
of immature invariant natural killer T cells accentuated of inflammation, has been identified as a dominant
disease severity in a model of crescentic glomerulo­ autoantigen that drives clonal B-cell selection in lupus
nephritis99. Thus, targeting CX3CL1/CX3CR1 pathway, tubulointerstitial inflammation, suggesting that vimen-
which regulates the activation of invariant natural killer tin might represent a therapeutic target for tubulointer-
T cells, might represent a promising treatment approach stitial lesions in lupus nephritis111. Anti-double-stranded
with fewer adverse effects than general immunosuppres- DNA (anti-dsDNA) antibodies can induce fibronectin
sive therapies (FIG. 3). Animal models studies have also secretion in proximal renal tubular epithelial cells, lead-
demonstrated that the IL‑23/IL‑17 pathway contributes ing to TGFβ activation and collagen synthesis, again
to renal injury in experimental glomerulonephritis100; highlighting potential therapeutic targets112. Thus, explo-
thus, blocking IL‑23 production and/or the subsequent rations into the pathogenesis of tubulointerstitial lesions
recruitment of IL‑17A‑producing effector γδT cell-­ in lupus nephritis will likely identify targeted therapeutic
receptor-expressing CD3+CD4−CD8− NK1.1 T cells, interventions for future study 113.
which are thought to have a role in crescent formation, This point is illustrated by a study that used novel
might also prove beneficial in limiting glomerular dam- computational approaches to quantify the expression
age in part through limiting the recruitment of myeloid of apoptosis regulators in infiltrating lymphocytes in
cells. Cell lineage-tracing studies indicate that podocytes biopsy samples from patients with lupus nephritis and
and parietal epithelial cells also participate in crescent tubulointerstital inflammation. The apoptosis regulator,
formation101,102. BCL‑2, was frequently expressed in these biopsy sam-
Importantly, in one study of 10 patients with ples, indicating a role for dysregulated apoptosis in the
lupus nephritis and glomerular crescents, all patients immune response. Moreover, treatment of NZB/WF1
were posi­t ive for serum ANCA and five had anti-­ lupus nephritis mice with a selective oral inhibitor of
myeloperoxidase (MPO) antibodies103. Similarly, another BCL‑2 prevented the development of tubulointerstitial
study reported that 10 of 33 Chinese patients with lupus inflammation114.
nephritis and crescents in ≥50% glomeruli were ANCA-
positive and seven had MPO-ANCA51. In fact, one study Renal vascular lesions. Vascular lesions are a common
reported a correlation between the serum ANCA sta- feature of SLE, and renal vasculopathy might repre-
tus and the presence of crescents in patients with lupus sent an extreme form of such lesions. Key pathogenic
nephritis104, and others have reported an association mechanisms include endothelial cell activation and
between atypical perinuclear ANCA, such as anti-cathepsin dysfunction, and dysregulation of the immune system,
Perinuclear-ANCA
G antibodies, and the development of crescentic lupus particularly through immune complex-induced vascular
A category of ANCA originally
described on the basis of their nephritis105,106. These findings provide strong evidence inflammation and antiphospholipid antibody-induced
immunofluorescence patterns for a role of ANCA in crescentic lupus nephritis, and thrombosis74,115 (FIG. 2). In addition to antiphospholipid
around the nucleus. suggest that therapeutic approaches that are used to antibodies, renal TMA in lupus nephritis can be caused
treat vasculitis, including intensive plasmapheresis and by thrombotic thrombocytopenic purpura, malignant
In situ adaptive immunity
A process in which infiltrate
the new anti‑C5aR antibody, should be assessed in this hypertension, pregnancy, scleroderma or drugs.
organizes into well- subgroup of patients51,103,107,108 (FIGS 3,4). Interestingly, studies have demonstrated a strong
circumscribed aggregates of relationship between the intensity of glomerular C4d
B cells and T cells (in germinal Podocyte injury. Proteinuria is a common feature of lupus staining and the presence of renal microthrombi in lupus
centres). These germinal
nephritis and reflects podocyte injury. Furthermore, the nephritis116,117. This finding suggests a role for the classical
centres contain follicular
dendritic cells and enable presence of nephrotic-range proteinuria in patients with complement pathway in lupus nephritis-­associated renal
intrarenal B cells to undergo lupus membranous nephropathy might represent the TMA, similar to the mechanisms involved in antiphos-
clonal expansion and somatic presence of podocyte dysfunction. Decreased expres- pholipid antibody-induced thrombosis and humoral
hypermutation. sion of the slit diaphragm proteins, nephrin and podo- rejection in transplanted kidneys116,117. We have demon-
Plasmablast foci
cin, in kidneys of mice and patients with lupus nephritis strated that the presence of glomerular C4d deposits and
Plasmablast aggregates in suggests that immune-mediated processes contribute to decreased serum levels of the complement control pro-
an organ. disruption of the slit diaphragm109. In patients with lupus tein, factor H are associated with poor renal outcomes in

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 491


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Class I • Supportive therapy


and II • Immunosuppressive therapy (proteinuria >3g per day)

• Maintenance
• Initial treatment
Class III • Treatment Refractory Add-on rituximab or
• H+CYC
and IV • H+MMF cases calcineurin inhibitor
• H+MMF
Classical • H+AZA
subtypes
• Anti-proteinuric and anti-hypertensive medications
Class V
• Immunosuppressive therapy H+CYC/MMF/AZA (persistent nephrotic proteinuria)

• Supportive therapy
Pathological Class VI
• Immunosuppressive therapy (external manifestations)
classification
of lupus
nephritis
Crescentic • PE/MP • CCX-168? (CD88 inhibitor)
GN • Rituximab (anti-CD20 Abs) • Abatacept? (CD80 inhibitor)
• Eculizumab? (Anti-C5 Abs)
Tubulo-
interstitial • Rituximab? (Anti-CD20 Abs)
Additional lesions • Abatacept? (CD80 inhibitor)
subtypes
Renal • PE/MP
vascular TMA • Eculizumab? (Anti-C5 Abs)
lesions • Thrombomodulin?

Podocyte • Abatacept? (CD80 inhibitor)


injury • Rituximab? (Anti-CD20 Abs)
• CNIs

Figure 4 | Proposed treatment algorithm for various pathological subtypes of lupus nephritis. Nature We propose
Reviews that the
| Nephrology
choice of treatment should depend on the pathological class of lupus nephritis as defined by the International Society of
Nephrology/Renal Pathology Society (ISN/RPS) classification system, and the presence of additional pathogenic subtypes
described in this Review. The treatment of ISN/RPS class I–VI lupus nephritis should follow basic therapeutic strategies
according to the three major guidelines1–3. For the treatment of the additional subtypes based on the various
pathogenesis, including crescentic lupus nephritis, podocyte injury, renal vascular lesions and tubulointerstitial lesions,
a series of therapeutic intervention recommendations are proposed. For patients with crescentic lupus nephritis (which
we define as the presence of crescents in ≥50% of glomeruli), we recommend first treating with plasma exchange (PE)
and methlyprednisolone pulse (MP), combined with cyclophosphamide or rituximab. Additional treatment options that
require further investigation include anti-complement agents (CCX‑168 and eculizumab) and the CD80 inhibitor
abatacept. Patients with severe tubulointerstitial lesions might be treated with rituximab or abatacept, although quality
clinical trials are lacking. For patients with renal vascular changes, especially those with thrombotic microangiopathy
(TMA), we suggest PE and MP as the first choice therapy. Eculizumab or recombinant thrombomodulin might also have
potential but require further exploration. Rituximab or abatacept might have a role in the treatment of patients with
prominent podocyte injury but further evidence of efficacy is required. In particular, the Kidney Disease: Improving Global
Outcome guideline suggests that steroids and calcineurin inhibitors (CNIs) should be used in lupus podocytopathy2.
Abs, antibodies; AZA, azathioprine; CYC, cyclophosphamide; GN, glomerulonephritis; H, corticosteroids;
MMF, mycophenolate mofetil.

patients with lupus nephritis and renal TMA, suggesting B cell-directed therapies. B cells have a critical role in
that activation of both the classical and alterative path- the pathogenesis of SLE beyond antibody production,
ways of complement might be involved in the develop- and B cell-depletion therapy has remained an attrac-
ment of TMA in these patients71,116,117. In the past few tive therapeutic option for nearly 10 years. Rituximab
years reports have emerged describing the successful use and ocrelizumab are humanized chimeric monoclonal
of the anti‑C5 monoclonal antibody, eculizumab, and anti‑CD20 antibodies that deplete autoreactive B cells
the anti-inflammatory, anticoagulant agent recombinant and thereby have the potential to attenuate the pro-
human soluble thrombo­modulin, in patients with lupus duction of ­autoantibodies that contribute to disease
­nephritis-­associated TMA and diffuse proliferative manifestations.
lupus nephritis resistant to conventional therapy, sug- The EXPLORER study of patients with class III or
gesting that direct targeting of the underlying pathogenic IV lupus nephritis, however, failed to demonstrate a
­mechanism is a promising approach118–121 (FIGS 3,4). benefit of adding rituximab to immunosuppressant
and cortico­steroid therapy 12. The subsequent LUNAR
Advances in novel therapies study of 144 patients with class III or IV lupus nephri-
Improved understanding of the pathogenesis of different tis showed that the addition of rituximab to MMF and
lupus nephritis subtypes suggests that various novel bio- corticosteroids led to greater reductions in levels of
logic agents and small molecules might be appropriate anti-dsDNA and in levels of the complement compo-
therapies for specific types of lupus nephritis. nents C3 and C4, but did not improve clinical outcomes

492 | AUGUST 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

after 1 year of treatment 122. The BELONG trial showed A systematic review of 2,004 patients with SLE pro-
a numerically but not statistically significant difference vides further support for targeting BAFF to induce or
in renal response rate with addition of ocrelizumab to ­maintain a renal response132.
standard therapy; however, this study was stopped early Neither rituximab nor belimumab deplete antibody-­
owing to a higher rate of serious infections123. Although producing plasma cells. Thus, inhibition of anti-
anti‑B‑cell therapies will eventually decrease inflamma- body production by plasma cells by the proteasome
tion by abrogating the generation of immune complexes inhibitor, bortezomib, remains a promising therapeutic
and intrarenal tertiary lymphoid organs, the failure of the strategy as has been demonstrated in mouse models of
above trials indicate that the effects of B‑cell depletion lupus nephritis133–135.
are not immediate nor direct and that anti‑B‑cell therapy
might be more effective for maintenance of remission T‑cell-directed therapies. Abatacept is a c­ o-stimulatory
than for induction of remission in patients with lupus inhibitor that targets B7‑1 (CD80) on the surface of
nephritis12,122,123. Contributing features that might have dendritic cells and B cells, blocking co-stimulatory
led to the failure of anti‑B‑cell therapy to demonstrate interaction between B7‑1 or B7‑2 and CD28 on T cells.
superiority over standard treatment regimens probably Unfortunately, abatacept as an add‑on to low-dose
also include the concomitant use of high-dose steroids, cyclophosphamide or MMF failed to improve complete
the use of stringent and nonorgan-specific criteria for renal response rates in clinical trials136–138. The failure of
clinical response, short follow‑up periods, small sample these trials might be due to the strict definitions of renal
sizes and a lack of an effect on critical non-circulating response; a revised global multi-centre trial of abatacept
pools of long-lived B cells. Nevertheless, the trials con- in lupus nephritis is ongoing 139.
firmed the safety of repeated treatment with rituximab124.
A small study of patients with severe lupus nephritis with Other therapeutics under development. Various other
mean follow‑up of nearly 4 years suggests that rituximab anti-inflammatory and/or immune regulatory agents are
shows promise as part of an intensified induction ther- under development and warrant investigation in lupus
apy protocol for patients who want to avoid or minimize nephritis140 (FIG. 3). Allogeneic mesenchymal stem cell
immunosuppressive maintenance therapy and oral ster- transplantation was used to successfully treat a small
oids125. The ongoing RITUXILUP study aims to demon- group of patients with lupus nephritis who were refrac-
strate whether the addition of rituximab to MMF therapy tory to conventional therapies141. Other therapies, such
is useful in treating lupus nephritis flares and whether as immuno-adsorption of anti-dsDNA antibodies and
it has a long lasting steroid-sparing beneficial effect 126. immuno-adsorption with a C1q column, have been
Preliminary findings suggest that rituximab can ena- reported to be useful in SLE and might also be useful
ble oral steroids to be safely avoided in the treatment of for the treatment of lupus nephritis142–145.
lupus nephritis127. Ongoing studies with obinutuzumab128 As our understanding of the pathogenic mechanisms
and ofatumumab129 — monoclonal anti‑CD20 antibodies of lupus nephritis continues to expand, more accurate
with greater capacity for B‑cell depletion than rituximab therapeutic targets will undoubtedly be identified (FIG. 4).
— are in progress130, and along with RITUXILUP, will
hopefully answer the remaining questions regarding the Conclusions
efficacy of anti‑B‑cell therapy in lupus nephritis. Although the 2003 ISN/RPS lupus nephritis classifi-
Of note, as B cells become depleted in response to cation system has gained international recognition,
anti‑CD20 antibodies, levels of B‑cell-activating factor intriguing controversies require further exploration
(BAFF; also known as TNFSF13B or BLyS) increase, and new insights into disease pathogenesis need to be
which might increase the generation of new autore- incorporated16. The identification of lesions — namely
active B cells, inhibit B-cell apoptosis and stimulate crescentic glomerulonephritis, podocyte injury, vascu-
the differentiation of B cells into immunoglobulin-­ lopathy and tubulointerstitial changes — that were not
producing plasma cells (FIG. 2). A phase III trial of beli- included in the original classification but are increasingly
mumab, a monoclonal antibody that neutralizes BAFF, recognized as being important in their contribution
in patients with SLE and renal involvement showed to the pathogenesis of disease, should be incorporated
that patients who received belimumab tended to have into the classification to improve its clinical value and aid
fewer renal flares and significantly greater reductions therapeutic decision making. We propose that targeting
in proteinuria than those treated with placebo, pro- these pathogenic pathways might enable a more person-
viding support for the use of B cell–directed therapies alized approach to the treatment of disease and lead to
in maintenance protocols for lupus nephritis131 (FIG. 3). improved outcomes for patients with lupus nephritis.

1. Bertsias, G. K. et al. Joint European League Against 3. Hahn, B. H. et al. American College of Rheumatology 6. Gatto, M. et al. Success and failure of biological
Rheumatism and European Renal Association– guidelines for screening, treatment, and management treatment in systemic lupus erythematosus:
European Dialysis and Transplant Association (EULAR/ of lupus nephritis. Arthritis Care Res. (Hoboken) 64, a critical analysis. J. Autoimmun. 74, 94–105
ERA–EDTA) recommendations for the management of 797–808 (2012). (2016).
adult and paediatric lupus nephritis. Ann. Rheum. Dis. 4. Weening, J. J. et al. The classification of 7. Narain, S. & Furie, R. Update on clinical trials in
71, 1771–1782 (2012). glomerulonephritis in systemic lupus erythematosus systemic lupus erythematosus. Curr. Opin. Rheumatol.
2. Kidney Disease: Improving Global Outcomes (KDIGO) revisited. Kidney Int. 65, 521–530 (2004). 28, 477–487 (2016).
Glomerulonephritis Work Group. KDIGO clinical 5. Anders, H. J. & Rovin, B. A pathophysiology-based 8. Parikh, S. V. & Rovin, B. H. Current and emerging
practice guideline for glomerulonephritis. Kidney Int. approach to the diagnosis and treatment of lupus therapies for lupus nephritis. J. Am. Soc. Nephrol. 27,
Suppl. 2, 139–274 (2012). nephritis. Kidney Int. 90, 493–501 (2016). 2929–2939 (2016).

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 493


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

9. Zhang, L. et al. Long-term outcomes of end-stage 34. Lv, J. et al. Evaluation of the Oxford classification of 59. Rezende, G. M. et al. Podocyte injury in pure
kidney disease for patients with lupus nephritis. IgA nephropathy: a systematic review and meta- membranous and proliferative lupus nephritis: distinct
Kidney Int. 89, 1337–1345 (2016). analysis. Am. J. Kidney Dis. 62, 891–899 (2013). underlying mechanisms of proteinuria? Lupus 23,
10. Appel, G. B. et al. Mycophenolate mofetil versus 35. Zeng, C. H. et al. A multicenter application and 255–262 (2014).
cyclophosphamide for induction treatment of lupus evaluation of the Oxford classification of IgA 60. Wang, Y., Yu, F., Song, D., Wang, S. X. & Zhao, M. H.
nephritis. J. Am. Soc. Nephrol. 20, 1103–1112 nephropathy in adult chinese patients. Am. J. Kidney Podocyte involvement in lupus nephritis based on the
(2009). Dis. 60, 812–820 (2012). 2003 ISN/RPS system: a large cohort study from a
11. Zavada, J. et al. Extended follow‑up of the CYCLOFA- 36. Shima, Y. et al. Validity of the Oxford classification of single centre. Rheumatology (Oxford) 53, 1235–1244
LUNE trial comparing two sequential induction and IgA nephropathy in children. Pediatr. Nephrol. 27, (2014).
maintenance treatment regimens for proliferative 783–792 (2012). 61. Yu, F. et al. Tubulointerstitial lesions of patients with
lupus nephritis based either on cyclophosphamide 37. Shi, S. F. et al. Pathologic predictors of renal outcome lupus nephritis classified by the 2003 International
oron cyclosporine A. Lupus 23, 69–74 (2014). and therapeutic efficacy in IgA nephropathy: validation Society of Nephrology and Renal Pathology Society
12. Rovin, B. H. et al. Efficacy and safety of rituximab in of the Oxford classification. Clin. J. Am. Soc. Nephrol. system. Kidney Int. 77, 820–829 (2010).
patients with active proliferative lupus nephritis: the 6, 2175–2184 (2011). 62. Hsieh, C. et al. Predicting outcomes of lupus nephritis
Lupus Nephritis Assessment with Rituximab study. 38. Kang, S. H. et al. The Oxford classification as a with tubulointerstitial inflammation and scarring.
Arthritis Rheum. 64, 1215–1226 (2012). predictor of prognosis in patients with IgA Arthritis Care Res. (Hoboken) 63, 865–874 (2011).
13. Wofsy, D., Hillson, J. L. & Diamond, B. Comparison nephropathy. Nephrol. Dial. Transplant. 27, 252–258 63. Alsuwaida, A. O. Interstitial inflammation and long-
of alternative primary outcome measures for use in (2012). term renal outcomes in lupus nephritis. Lupus 22,
lupus nephritis clinical trials. Arthritis Rheum. 65, 39. Working Group of the International IgA Nephropathy 1446–1454 (2013).
1586–1591 (2013). Network and the Renal Pathology Society et al. The 64. Hill, G. S., Delahousse, M., Nochy, D., Mandet, C. &
14. Banchereau, R. et al. Personalized immunomonitoring Oxford IgA nephropathy clinicopathological Bariety, J. Proteinuria and tubulointerstitial lesions in
uncovers molecular networks that stratify lupus classification is valid for children as well as adults. lupus nephritis. Kidney Int. 60, 1893–1903 (2001).
patients. Cell 165, 551–565 (2016). Kidney Int. 77, 921–927 (2010). 65. Hill, G. S. et al. Predictive power of the second renal
15. Haas, M., Rastaldi, M. P. & Fervenza, F. C. Histologic 40. Cattran, D. C. et al. The Oxford classification of IgA biopsy in lupus nephritis: significance of macrophages.
classification of glomerular diseases: clinicopathologic nephropathy: rationale, clinicopathological Kidney Int. 59, 304–316 (2001).
correlations, limitations exposed by validation studies, correlations, and classification. Kidney Int. 76, 66. Zappitelli, M., Duffy, C. M., Bernard, C. & Gupta, I. R.
and suggestions for modification. Kidney Int. 85, 534–545 (2009). Evaluation of activity, chronicity and tubulointerstitial
779–793 (2014). 41. Roberts, I. S. et al. The Oxford classification of IgA indices for childhood lupus nephritis. Pediatr. Nephrol.
16. Wilhelmus, S. et al. The revisited classification of GN nephropathy: pathology definitions, correlations, and 23, 83–91 (2008).
in SLE at 10 years: time to re‑evaluate histopathologic reproducibility. Kidney Int. 76, 546–556 (2009). 67. Zhang, X. et al. A composite urine biomarker reflects
lesions. J. Am. Soc. Nephrol. 26, 2938–2946 (2015). 42. Najafi, C. C. et al. Significance of histologic patterns of interstitial inflammation in lupus nephritis kidney
17. Parikh, S. V., Alvarado, A., Malvar, A. & Rovin, B. H. glomerular injury upon long-term prognosis in severe biopsies. Kidney Int. 81, 401–406 (2012).
The kidney biopsy in lupus nephritis: past, present, lupus glomerulonephritis. Kidney Int. 59, 2156–2163 68. Appel, G. B., Pirani, C. L. & D’Agati, V. Renal vascular
and future. Semin. Nephrol. 35, 465–477 (2015). (2001). complications of systemic lupus erythematosus.
18. Parikh, S. V., Ayoub, I. & Rovin, B. H. The kidney 43. Mittal, B., Hurwitz, S., Rennke, H. & Singh, A. K. J. Am. Soc. Nephrol. 4, 1499–1515 (1994).
biopsy in lupus nephritis: time to move beyond New subcategories of class IV lupus nephritis: are 69. Wu, L. H. et al. Inclusion of renal vascular lesions
histology. Nephrol. Dial. Transplant. 30, 3–6 (2015). there clinical, histologic, and outcome differences? in the 2003 ISN/RPS system for classifying lupus
19. Malvar, A. et al. Histologic versus clinical remission Am. J. Kidney Dis. 44, 1050–1059 (2004). nephritis improves renal outcome predictions.
in proliferative lupus nephritis. Nephrol. Dial. 44. Hill, G. S., Delahousse, M., Nochy, D. & Bariety, J. Kidney Int. 83, 715–723 (2013).
Transplant. http:dx.doi.org/10.1093/ndt/gfv296 Class IV‑S versus class IV‑G lupus nephritis: clinical and 70. Barber, C. et al. Evaluation of clinical outcomes and
(2015). morphologic differences suggesting different renal vascular pathology among patients with lupus.
20. Rovin, B. H., Parikh, S. V. & Alvarado, A. The kidney pathogenesis. Kidney Int. 68, 2288–2297 (2005). Clin. J. Am. Soc. Nephrol. 7, 757–764 (2012).
biopsy in lupus nephritis: is it still relevant? 45. Yu, F. et al. Class IV‑G and IV‑S lupus nephritis in 71. Song, D. et al. The spectrum of renal thrombotic
Rheum. Dis. Clin. North Am. 40, 537–552 (2014). Chinese patients: a large cohort study from a single microangiopathy in lupus nephritis. Arthritis Res. Ther.
21. Zhu, D., Qu, Z., Tan, Y., Yu, F. & Zhao, M. H. Acute center. Lupus 18, 1073–1081 (2009). 15, R12 (2013).
kidney injury in Chinese patients with lupus nephritis: 46. Schwartz, M. M., Korbet, S. M. & Lewis, E. J. 72. Yu, F., Tan, Y. & Zhao, M. H. Lupus nephritis combined
a large cohort study from a single center. Lupus 20, The prognosis and pathogenesis of severe lupus with renal injury due to thrombotic thrombocytopaenic
1557–1565 (2011). glomerulonephritis. Nephrol. Dial. Transplant. 23, purpura-haemolytic uraemic syndrome. Nephrol. Dial.
22. Churg, J., Bernstein, J. & Classock, R. J. Renal 1298–1306 (2008). Transplant. 25, 145–152 (2010).
Disease: Classification and Atlas of Glomerular 47. Haring, C. M., Rietveld, A., van den Brand, J. A. & 73. Chu, H., Wu, L. H., Song, D., Yu, F. & Zhao, M. H.
Disease 2nd edn (Igaku-Shoin, 1995). Berden, J. H. Segmental and global subclasses of Noninflammatory necrotizing vasculopathy in lupus
23. Furness, P. N. & Taub, N. Interobserver reproducibility class IV lupus nephritis have similar renal outcomes. nephritis: a single-center experience. Lupus 23,
and application of the ISN/RPS classification of lupus J. Am. Soc. Nephrol. 23, 149–154 (2012). 20–30 (2014).
nephritis‑a UK‑wide study. Am. J. Surg. Pathol. 30, 48. Churg, J. & Sobin, L. Renal Disease: Classification and 74. Tan, Y., Yu, F. & Liu, G. Diverse vascular lesions in
1030–1035 (2006). Atlas of Glomerular Disease 1st edn (Igaku-Shoin, systemic lupus erythematosus and clinical
24. Grootscholten, C. et al. Interobserver agreement of 1982). implications. Curr. Opin. Nephrol. Hypertens. 23,
scoring of histopathological characteristics and 49. Hill, G. S. et al. A new morphologic index for the 218–223 (2014).
classification of lupus nephritis. Nephrol. Dial. evaluation of renal biopsies in lupus nephritis. 75. Huang, J. et al. Renal interstitial arteriosclerotic
Transplant. 23, 223–230 (2008). Kidney Int. 58, 1160–1173 (2000). lesions in lupus nephritis patients: a cohort study
25. Yokoyama, H. et al. The outcome and a new ISN/RPS 50. Sumethkul, V., Chalermsanyakorn, P., from China. PLoS ONE 10, e0141547 (2015).
2003 classification of lupus nephritis in Japanese. Changsirikulchai, S. & Radinahamed, P. Lupus 76. Liu, Z. et al. Multitarget therapy for induction
Kidney Int. 66, 2382–2388 (2004). nephritis: a challenging cause of rapidly progressive treatment of lupus nephritis: a randomized trial.
26. Chan, T. M. Histological reclassification of lupus crescentic glomerulonephritis. Lupus 9, 424–428 Ann. Intern. Med. 162, 18–26 (2015).
nephritis. Curr. Opin. Nephrol. Hypertens. 14, (2000). 77. Allison, S. J. Lupus nephritis: multitarget induction
561–566 (2005). 51. Yu, F. et al. Clinicopathological characteristics and therapy for LN. Nat. Rev. Nephrol. 11, 3 (2015).
27. Kojo, S. et al. Clinical usefulness of a prognostic score outcomes of patients with crescentic lupus nephritis. 78. Anders, H. J. & Fogo, A. B. Immunopathology of lupus
in histological analysis of renal biopsy in patients with Kidney Int. 76, 307–317 (2009). nephritis. Semin. Immunopathol. 36, 443–459
lupus nephritis. J. Rheumatol. 36, 2218–2223 52. Horino, T. et al. Minimal change nephrotic syndrome (2014).
(2009). associated with systemic lupus erythematosus. 79. Bruschi, M. et al. Glomerular autoimmune
28. Markowitz, G. S. & D’Agati, V. D. The ISN/RPS 2003 Nephrol. Dial. Transplant. 21, 230 (2006). multicomponents of human lupus nephritis in vivo:
classification of lupus nephritis: an assessment at 53. Hertig, A. et al. SLE and idiopathic nephrotic alpha-enolase and annexin AI. J. Am. Soc. Nephrol.
3 years. Kidney Int. 71, 491–495 (2007). syndrome: coincidence or not? Am. J. Kidney Dis. 40, 25, 2483–2498 (2014).
29. Markowitz, G. S. & D’Agati, V. D. Classification of 1179–1184 (2002). 80. Chan, T. M. Treatment of severe lupus nephritis: the
lupus nephritis. Curr. Opin. Nephrol. Hypertens. 18, 54. Desai, N., Cimbaluk, D., Lewis, E. J. & Whittier, W. L. new horizon. Nat. Rev. Nephrol. 11, 46–61 (2015).
220–225 (2009). Proteinuria in membranous lupus nephritis: the 81. Lech, M. & Anders, H. J. The pathogenesis of lupus
30. Schwartz, M. M. The pathology of lupus nephritis. pathology is in the podocyte. Lupus 22, 461–468 nephritis. J. Am. Soc. Nephrol. 24, 1357–1366
Semin. Nephrol. 27, 22–34 (2007). (2013). (2013).
31. Schwartz, M. M., Korbet, S. M., Katz, R. S. & 55. Kraft, S. W., Schwartz, M. M., Korbet, S. M. & 82. Lorenz, G., Desai, J. & Anders, H. J. Lupus nephritis:
Lewis, E. J. Evidence of concurrent Lewis, E. J. Glomerular podocytopathy in patients with update on mechanisms of systemic autoimmunity and
immunopathological mechanisms determining the systemic lupus erythematosus. J. Am. Soc. Nephrol. kidney immunopathology. Curr. Opin. Nephrol.
pathology of severe lupus nephritis. Lupus 18, 16, 175–179 (2005). Hypertens. 23, 211–217 (2014).
149–158 (2009). 56. Shea-Simonds, P., Cairns, T. D., Roufosse, C., Cook, T. 83. Mohan, C. & Putterman, C. Genetics and pathogenesis
32. Wilhelmus, S. et al. Interobserver agreement on & Vyse, T. J. Lupus podocytopathy. Rheumatology of systemic lupus erythematosus and lupus nephritis.
histopathological lesions in class III or IV lupus (Oxford) 48, 1616–1618 (2009). Nat. Rev. Nephrol. 11, 329–341 (2015).
nephritis. Clin. J. Am. Soc. Nephrol. 10, 47–53 57. Salvatore, S. P. et al. Collapsing glomerulopathy in 19 84. Nowling, T. K. & Gilkeson, G. S. Mechanisms of tissue
(2015). patients with systemic lupus erythematosus or lupus- injury in lupus nephritis. Arthritis Res. Ther. 13, 250
33. Coppo, R. et al. VALIGA study of the ERA–EDTA like disease. Clin. J. Am. Soc. Nephrol. 7, 914–925 (2011).
Immunonephrology Working Group. Validation of the (2012). 85. Bekar, K. W. et al. Prolonged effects of short-term
Oxford classification of IgA nephropathy in cohorts with 58. Hu, W. et al. Clinical–morphological features and anti‑CD20 B cell depletion therapy in murine systemic
different presentations and treatments. Kidney Int. 86, outcomes of lupus podocytopathy. Clin. J. Am. Soc. lupus erythematosus. Arthritis Rheum. 62,
828–836 (2014). Nephrol. 11, 585–592 (2016). 2443–2457 (2010).

494 | AUGUST 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

86. Ramanujam, M. et al. Selective blockade of BAFF stages of lupus nephritis and correlate with disease 130. Reddy, V., Dahal, L. N., Cragg, M. S. & Leandro, M.
for the prevention and treatment of systemic lupus histology. Lupus 20, 781–791 (2011). Optimising B‑cell depletion in autoimmune disease:
erythematosus nephritis in NZM2410 mice. 110. Chang, A. et al. In situ B cell-mediated immune is obinutuzumab the answer? Drug Discov. Today 21,
Arthritis Rheum. 62, 1457–1468 (2010). responses and tubulointerstitial inflammation in 1330–1338 (2016).
87. Chan, T. M. et al. Efficacy of mycophenolate mofetil human lupus nephritis. J. Immunol. 186, 1849–1860 131. Dooley, M. A. et al. Effect of belimumab treatment on
in patients with diffuse proliferative lupus nephritis. (2011). renal outcomes: results from the phase 3 belimumab
Hong Kong–Guangzhou Nephrology Study Group. 111. Kinloch, A. J. et al. Vimentin is a dominant target of clinical trials in patients with SLE. Lupus 22, 63–72
N. Engl. J. Med. 343, 1156–1162 (2000). in situ humoral immunity in human lupus (2013).
88. Ginzler, E. M. et al. Mycophenolate mofetil or tubulointerstitial nephritis. Arthritis Rheumatol. 66, 132. Sciascia, S. et al. Efficacy of belimumab on renal
intravenous cyclophosphamide for lupus nephritis. 3359–3370 (2014). outcomes in patients with systemic lupus
N. Engl. J. Med. 353, 2219–2228 (2005). 112. Yung, S. et al. Anti-dsDNA antibody induces soluble erythematosus: a systematic review. Autoimmun. Rev.
89. Hannah, J., Casian, A. & D’Cruz, D. Tacrolimus use in fibronectin secretion by proximal renal tubular 16, 287–293 (2017).
lupus nephritis: a systematic review and meta-analysis. epithelial cells and downstream increase of TGF‑beta1 133. Ichikawa, H. T. et al. Beneficial effect of novel
Autoimmun. Rev. 15, 93–101 (2016). and collagen synthesis. J. Autoimmun. 58, 111–122 proteasome inhibitors in murine lupus via dual
90. Chen, W. et al. Short-term outcomes of induction (2015). inhibition of type I interferon and autoantibody-
therapy with tacrolimus versus cyclophosphamide 113. Yung, S. & Chan, T. M. Molecular and immunological secreting cells. Arthritis Rheum. 64, 493–503 (2012).
for active lupus nephritis: a multicenter randomized basis of tubulo-interstitial injury in lupus nephritis: 134. Neubert, K. et al. The proteasome inhibitor
clinical trial. Am. J. Kidney Dis. 57, 235–244 a comprehensive review. Clin. Rev. Allergy Immunol. bortezomib depletes plasma cells and protects mice
(2011). 52, 149–163 (2016). with lupus-like disease from nephritis. Nat. Med. 14,
91. Li, X. et al. Mycophenolate mofetil or tacrolimus 114. Ko, K. et al. BCL‑2 as a therapeutic target in human 748–755 (2008).
compared with intravenous cyclophosphamide in the tubulointerstitial inflammation. Arthritis Rheumatol. 135. Khodadadi, L. et al. Bortezomib plus continuous B cell
induction treatment for active lupus nephritis. 68, 2740–2751 (2016). depletion results in sustained plasma cell depletion
Nephrol. Dial. Transplant. 27, 1467–1472 (2012). 115. Frieri, M. Accelerated atherosclerosis in systemic and amelioration of lupus nephritis in NZB/W F1 mice.
92. US National Library of Medicine. ClinicalTrials.gov lupus erythematosus: role of proinflammatory PLoS ONE 10, e0135081 (2015).
https://clinicaltrials.gov/ct2/show/NCT02141672 cytokines and therapeutic approaches. Curr. Allergy 136. Furie, R. et al. Efficacy and safety of abatacept in lupus
(2017). Asthma Rep. 12, 25–32 (2012). nephritis: a twelve-month, randomized, double-blind
93. Corapi, K. M., Dooley, M. A., Pendergraft, W. F. III. 116. Chua, J. S. et al. Complement factor C4d is a common study. Arthritis Rheumatol. 66, 379–389 (2014).
Comparison and evaluation of lupus nephritis denominator in thrombotic microangiopathy. 137. Fragoso-Loyo, H. E. & Dooley, M. A. Efficacy of the
response criteria in lupus activity indices andclinical J. Am. Soc. Nephrol. 26, 2239–2247 (2015). euro-lupus nephritis regimen combined with abatacept
trials. Arthritis Res. Ther. 17, 110 (2015). 117. Cohen, D. et al. Potential for glomerular C4d as an versus placebo in a North American lupus nephritis
94. Mok, C. C. Membranous nephropathy in systemic indicator of thrombotic microangiopathy in lupus population. Ann. Rheum. Dis. 73 (Suppl. 2), 77–78
lupus erythematosus: a therapeutic enigma. nephritis. Arthritis Rheum. 58, 2460–2469 (2008). (2014).
Nat. Rev. Nephrol. 5, 212–220 (2009). 118. El‑Husseini, A. et al. Thrombotic microangiopathy in 138. Group, A. T. Treatment of lupus nephritis with
95. Yap, D. Y. et al. Long-term data on tacrolimus systemic lupus erythematosus: efficacy of eculizumab. abatacept: the Abatacept and Cyclophosphamide
treatment in lupus nephritis. Rheumatology (Oxford) Am. J. Kidney Dis. 65, 127–130 (2015). Combination Efficacy and Safety Study.
53, 2232–2237 (2014). 119. Coppo, R. et al. Dramatic effects of eculizumab in a Arthritis Rheumatol. 66, 3096–3104 (2014).
96. Beck, L. H. Jr & Salant, D. J. Treatment of child with diffuse proliferative lupus nephritis resistant 139. US National Library of Medicine. ClinicalTrials.gov
membranous lupus nephritis: where are we now? to conventional therapy. Pediatr. Nephrol. 30, http://www.clinicaltrials.gov/ct2/show/
J. Am. Soc. Nephrol. 20, 690–691 (2009). 167–172 (2015). NCT01714817?term=NCT01714817&rank=1
97. Austin, H. A. III, Illei, G. G., Braun, M. J. & Balow, J. E. 120. Tonooka, K., Ito, H., Shibata, T. & Ozaki, S. (2017).
Randomized, controlled trial of prednisone, Recombinant human soluble thrombomodulin for 140. Rovin, B. H. & Parikh, S. V. Lupus nephritis: the
cyclophosphamide, and cyclosporine in lupus treatment of thrombotic microangiopathy associated evolving role of novel therapeutics. Am. J. Kidney Dis.
membranous nephropathy. J. Am. Soc. Nephrol. 20, with lupus nephritis. J. Rheumatol. 39, 1766–1767 63, 677–690 (2014).
901–911 (2009). (2012). 141. Gu, F. et al. Allogeneic mesenchymal stem cell
98. Migliorini, A. et al. The antiviral cytokines IFN-alpha 121. Leffler, J., Bengtsson, A. A. & Blom, A. M. The transplantation for lupus nephritis patients refractory
and IFN-beta modulate parietal epithelial cells and complement system in systemic lupus erythematosus: to conventional therapy. Clin. Rheumatol. 33,
promote podocyte loss: implications for IFN toxicity, an update. Ann. Rheum. Dis. 73, 1601–1606 (2014). 1611–1619 (2014).
viral glomerulonephritis, and glomerular regeneration. 122. Merrill, J. T. et al. Efficacy and safety of rituximab in 142. Olson, S. W. et al. Elevated subclinical double-stranded
Am. J. Pathol. 183, 431–440 (2013). moderately-to‑severely active systemic lupus DNA antibodies and future proliferative lupus nephritis.
99. Mesnard, L. et al. Invariant natural killer T cells and erythematosus: the randomized, double-blind, Clin. J. Am. Soc. Nephrol. 8, 1702–1708 (2013).
TGF-beta attenuate anti-GBM glomerulonephritis. phase II/III systemic lupus erythematosus evaluation 143. Orbai, A. M. et al. Anti‑C1q antibodies in systemic
J. Am. Soc. Nephrol. 20, 1282–1292 (2009). of  rituximab trial. Arthritis Rheum. 62, 222–233 lupus erythematosus. Lupus 24, 42–49 (2015).
100. Paust, H. J. et al. The IL‑23/Th17 axis contributes to (2010). 144. Pfueller, B., Wolbart, K., Bruns, A., Burmester, G. R.
renal injury in experimental glomerulonephritis. 123. Mysler, E. F. et al. Efficacy and safety of ocrelizumab in & Hiepe, F. Successful treatment of patients with
J. Am. Soc. Nephrol. 20, 969–979 (2009). active proliferative lupus nephritis: results from a systemic lupus erythematosus by immunoadsorption
101. Smeets, B. et al. Tracing the origin of glomerular randomized, double-blind, phase III study. Arthritis with a C1q column: a pilot study. Arthritis Rheum. 44,
extracapillary lesions from parietal epithelial cells. Rheum. 65, 2368–2379 (2013). 1962–1963 (2001).
J. Am. Soc. Nephrol. 20, 2604–2615 (2009). 124. Reddy, V., Jayne, D., Close, D. & Isenberg, D. B‑Cell 145. Suzuki, K. The role of immunoadsorption using
102. Ohse, T. et al. De novo expression of podocyte depletion in SLE: clinical and trial experience with dextran-sulfate cellulose columns in the treatment
proteins in parietal epithelial cells during experimental rituximab and ocrelizumab and implications for study of systemic lupus erythematosus. Ther. Apher. 4,
glomerular disease. Am. J. Physiol. Renal Physiol. design. Arthritis Res. Ther. 15 (Suppl. 1), S2 (2013). 239–243 (2000).
298, F702–F711 (2010). 125. Roccatello, D. et al. A 4‑year observation in lupus
103. Nasr, S. H. et al. Necrotizing and crescentic lupus nephritis patients treated with an intensified Acknowledgements
nephritis with antineutrophil cytoplasmic antibody B‑lymphocyte depletion without immunosuppressive We gratefully acknowledge the contributions of Hong Chu,
seropositivity. Clin. J. Am. Soc. Nephrol. 3, 682–690 maintenance treatment — clinical response compared Ying Tan, Chen Wang and Su‑xia Wang, Peking University First
(2008). to literature and immunological re‑assessment. Hospital, Beijing, for collecting data discussed in this Review.
104. Lee, S. S. et al. Antilactoferrin antibody in systemic Autoimmun. Rev. 14, 1123–1130 (2015). This work is supported by grants of National Natural Science
lupus erythematosus. Br. J. Rheumatol. 31, 669–673 126. US National Library of Medicine. ClinicalTrials.gov Foundation of China to Innovation Research Group
(1992). http://www.clinicaltrials.gov/ct2/show/ (No. 81621092) and National Natural Science Foundation of
105. Chin, H. J. et al. Clinical implications of antineutrophil NCT01773616?term=NCT01773616&rank=1 China (No. 81470932, No.81500526, No. 81670640 and
cytoplasmic antibody test in lupus nephritis. (2017). No. 81670639).
Am. J. Nephrol. 20, 57–63 (2000). 127. Condon, M. B. et al. Prospective observational single-
106. Zhao, M. H., Liu, N., Zhang, Y. K. & Wang, H. Y. centre cohort study to evaluate the effectiveness of Author contributions
Antineutrophil cytoplasmic autoantibodies (ANCA) treating lupus nephritis with rituximab and All authors researched data for the article, provided substan-
and their target antigens in Chinese patients with mycophenolate mofetil but no oral steroids. tial contributions to discussions of its content, wrote the arti-
lupus nephritis. Nephrol. Dial. Transplant. 13, Ann. Rheum. Dis. 72, 1280–1286 (2013). cle, and reviewed and/or edited of the manuscript before
2821–2824 (1998). 128. US National Library of Medicine. ClinicalTrials.gov submission.
107. Xiao, H. et al. C5a receptor (CD88) blockade protects http://www.clinicaltrials.gov/ct2/show/
against MPO-ANCA GN. J. Am. Soc. Nephrol. 25, NCT02550652?term=NCT02550652&rank=1 Competing interests statement
225–231 (2014). (2017). R.G. consults for Genentech. The other authors declare no
108. US National Library of Medicine. ClinicalTrials.gov 129. Karageorgas, T. et al. Successful treatment of life- competing interests.
https://clinicaltrials.gov/ct2/show/NCT01363388 threatening autoimmune haemolytic anaemia with
(2016). ofatumumab in a patient with systemic lupus Publisher’s note
109. Perysinaki, G. S. et al. Podocyte main slit diaphragm erythematosus. Rheumatology (Oxford) 55, Springer Nature remains neutral with regard to jurisdictional
proteins, nephrin and podocin, are affected at early 2085–2087 (2016). claims in published maps and institutional affiliations.

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | AUGUST 2017 | 495


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

S-ar putea să vă placă și