You are on page 1of 17

REVIEWS

Treatment of membranous
nephropathy: time for a paradigm shift
Piero Ruggenenti1,2, Fernando C. Fervenza3 and Giuseppe Remuzzi1,2,4
Abstract | In patients with membranous nephropathy, alkylating agents (cyclophosphamide or
chlorambucil) alone or in combination with steroids achieve remission of nephrotic syndrome
more effectively than conservative treatment or steroids alone, but can cause myelotoxicity,
infections, and cancer. Calcineurin inhibitors can improve proteinuria, but are nephrotoxic.
Most patients relapse after treatment withdrawal and can become treatment dependent, which
increases the risk of nephrotoxicity. The discovery of nephritogenic autoantibodies against
podocyte M‑type phospholipase A2 receptor (PLA2R) and thrombospondin type‑1 domain-
containing protein 7A (THSD7A) antigens provides a clear pathophysiological rationale for
interventions that specifically target B‑cell lineages to prevent antibody production and
subepithelial deposition. The anti‑CD20 monoclonal antibody rituximab is safe and achieves
remission of proteinuria in approximately two-thirds of patients with membranous nephropathy.
In those with PLA2R-related disease, remission can be predicted by anti-PLA2R antibody depletion
and relapse by antibody re‑emergence into the circulation. Thus, integrated evaluation of
serology and proteinuria could guide identification of affected patients and treatment with
individually tailored protocols. Nonspecific and toxic immunosuppressive regimens will fall out
of use. B-cell modulation by rituximab and second-generation anti‑CD20 antibodies (or plasma
cell-targeted therapy in anti‑CD20 resistant forms of disease) will lead to a novel therapeutic
paradigm for patients with membranous nephropathy.

1
IRCCS – Istituto di Ricerche Membranous nephropathy is an immune-mediated dis- involves the use of medications that do not interfere with
Farmacologiche Mario Negri, ease that results from the deposition of IgG and com- disease-specific mechanisms but instead aims to correct
Centro Anna Maria Astori,
Science and Technology Park
plement components onto the subepithelial layer of the or ameliorate concomitant pathophysiological changes
Kilometro Rosso, glomerular capillary wall. The disease affects ~5–10 that might contribute to disease progression or the onset
Via Stezzano 87, patients per million population and is the leading cause of complications such as proteinuria, which is the strong-
24126 Bergamo, Italy. of nephrotic syndrome in adults1. Solid or haematological est predictor of disease outcome in membranous nephro­
2
Unit of Nephrology and
malignancies, systemic autoimmune diseases, infections pathy and most other chronic nephropathies3–5. Without
Dialysis, Azienda Socio-
Sanitaria Territoriale Papa or the chronic consumption of drugs such as gold salts, immunosuppression, approximately one-third of patients
Giovanni XXIII, Piazza OMS 1, NSAIDs and penicillamine seem to have a role in disease with membranous nephropathy and nephrotic syndrome
24128, Bergamo, Italy. pathogenesis in approximately one-third of cases, which (proteinuria >3.5 g per 24 h and/or hypoalbuminaemia)
3
Division of Nephrology and are usually classified as secondary forms of membranous will progress to end-stage renal disease (ESRD)6. By con-
Hypertension, Mayo Clinic,
200 First Street SW,
nephropathy. A majority of the remaining 70% of cases trast, the clinical condition of some of the remaining two-
Rochester, Minnesota, USA. are defined by the presence of specific nephritogenic thirds of patients will remain stable over time, whereas
4
Department of Biomedical autoantibodies and are usually described as primary others will achieve spontaneous remission of proteinuria
and Clinical Sciences, membranous nephropathy. The term idiopathic mem- with sustained stabilization of renal function7. The ina-
University of Milan, Via Festa
branous nephropathy should probably be restricted to bility of nephrologists to discriminate between progres-
del Perdono 7, 20122 Milan
Italy. cases in which autoantibodies cannot be identified. To sive and non-progressive disease has therefore fuelled
Correspondence to G.R.
avoid confusion, here we refer to primary and idiopathic lively debate about the optimal treatment of patients with
Giuseppe.remuzzi@ forms simply as membranous nephropathy 2. ­membranous nephropathy and nephrotic syndrome8.
marionegri.it Patients with membranous nephropathy and non-­ Following a brief discussion of the role of supportive
doi:10.1038/nrneph.2017.92 nephrotic proteinuria (<3.5 g per 24 h) have a good therapy and the risk:benefit profile of traditional, non-
Published online 3 Jul 2017 prognosis with supportive therapy. Supportive therapy specific immunosuppressive regimens, here we focus on

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 563


©
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 whenever feasible (although symptomatic hypo­


tension can be an insuperable barrier in normotensive
• Steroids and alkylating agents increase the rate of remission in patients with patients22,23) to improve glomerular barrier size selectiv-
membranous nephropathy but can cause diabetes mellitus, myelotoxicity, infections, ity, reduce proteinuria, and increase serum albumin ­levels
cancer, and other adverse events with consequent correction of hypercholesterolaemia
• Calcineurin inhibitors can reduce proteinuria in patients with membranous and other components of the nephrotic syndrome24,25.
nephropathy but are nephrotoxic, and these agents should be avoided in patients ACE inhibitors can also reduce serum triglyceride levels
with abnormal kidney function
by improving insulin sensitivity 24.
• The discovery of nephritogenic autoantibodies provided a clear rationale for The Remission Clinic programme, which employs
interventions specifically aimed at preventing formation of antigen–antibody
a multimodal intervention strategy that is titrated to
immunocomplexes with secondary complement activation, podocyte damage,
and proteinuria
urinary protein level and maximal RAS inhibition
(BOX 1), slowed the decline in glomerular filtration rate
• B cell-targeted therapy with rituximab is at least as effective as steroids and alkylating
agents in achieving remission in membranous nephropathy, and available evidence
and almost fully prevented progression to ESRD in
suggests that it is safer and better tolerated patients with nephrotic syndrome secondary to chronic
• In patients with anti-PLA2R-related membranous nephropathy and overt nephrotic
nephropathies, including membranous nephropathy 26–28.
syndrome, evaluation of serum autoantibody titre and albumin levels, as well as This finding is clinically relevant because, as mentioned
assessment of proteinuria could guide tailored therapy above, proteinuria is the strongest predictor of long-term
• Traditional immunosuppressive regimens will be replaced by specific, nontoxic agents renal outcome in patients with chronic kidney disease.
such as B cell-targeting monoclonal antibodies; modulation of B-cell immunity could Indeed, either complete or partial remission of protein-
lead to a novel therapeutic paradigm in membranous nephropathy uria protects from progression to ESRD, an effect that
correlates with duration of remission and is independent
of age, sex, and residual kidney function3. In this con-
novel and safer treatment approaches involving B cell-­ text, immunosuppressive therapy might be indicated
depleting monoclonal antibodies, such as rituximab for patients with membranous nephropathy who do not
or ofatumumab, which specifically target mechanisms achieve remission with supportive therapy alone or can-
involved in the production of autoantibodies that lead to not implement the Remission Clinic protocol because of
progressive structural damage and kidney dysfunction. symptomatic hypotension or hyperkalaemia25.
Finally, we discuss the potential role of rescue therapy
with agents that target plasma cells, which might prevent Traditional immunosuppressive therapy
antibody–antigen interactions and immune complex-­ Whether specific interventions (TABLE 1) improve disease
mediated complement activation. In the era of precision outcome compared with supportive therapy or placebo
medicine9, improved understanding of disease mech­ has not been established, as the trials that have been
anisms and the development of disease-specific medica- conducted were underpowered. In a systematic review
tions will hopefully pave the way to even safer and more that included 1,025 patients from 18 trials conducted up
effective individually tailored therapeutic options for to December 2003, glucocorticoids alone were found
patients with membranous nephropathy. not to affect proteinuria29. However, alkylating agents
(cyclophosphamide or chlorambucil) alone or in com-
Supportive therapy bination with steroids increased the rate of complete or
All patients with membranous nephropathy should partial remissions compared with supportive treatment
receive optimal supportive therapy to reduce pro- or steroids alone29. The treatment benefit was confined
teinuria 10. Treatments include renin–angiotensin to patients with serum creatinine level <1.1 mg/dl
system (RAS) inhibitors such as angiotensin-converting-­ (<97 μmol/l), that is, those patients who seldom progress
enzyme (ACE) inhibitors and angiotensin-receptor to ESRD, even without immunosuppression30. Moreover,
blockers (ARBs)5, which reduce protein ultrafiltration serious adverse events (SAEs) were much more frequent
by ameliorating glomerular hypertension and improving among patients receiving steroids or alkylating agents
glomerular barrier size selectivity11,12. Proteinuria usually than among other patients29. Cyclophosphamide is safer
decreases within 2 months of treatment 13. However, in than chlorambucil but results in more treatment with-
patients with severe proteinuria (that is, those who are drawals and hospitalizations than conservative ther-
at highest risk of disease progression), the decrease is apy 31,32. No treatment benefits, only adverse effects, were
seldom >30% from pretreatment values13,14. RAS inhibi- observed with ciclosporin31. In a large, randomized con-
tors therefore seem to be less nephroprotective in mem- trolled trial (RCT) conducted in the UK, steroids plus
branous nephropathy than in other proteinuric chronic chlorambucil were found to slow the loss of renal func-
nephropathies15–18. Combination therapy with aldoster- tion compared with supportive therapy alone in patients
one antagonists19 or NSAIDs20 does not add to the anti- with membranous nephropathy 33. However, the 3‑year
proteinuric effects of RAS inhibitors, and could increase follow‑up period was too short to evaluate whether
serum potassium levels. Combination therapy with ACE treatment also prevented ESRD. Moreover, prednisolone
inhibitors and ARBs is more effective at reducing pro- and chlorambucil combination therapy was associated
teinuria than ACE inhibitor or ARB monotherapy in with a nearly twofold higher incidence of SAEs com-
patients with chronic nephropathy not related to dia- pared with supportive therapy (2.8 events per patient
betes mellitus, including membranous nephropathy 21. versus 1.5 events per patient). Overall, 36 of the 56 events
Dual RAS blockade should therefore be implemented observed with specific treatment were thought possibly

564 | SEPTEMBER 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

or likely to be treatment-related. Events included two Box 1 | The Remission Clinic protocol*
malignancies, 28 cases of bone marrow toxicity, and
eight of impaired glucose tolerance. This event rate • Low sodium diet with or without diuretics and protein
exceeded the rates reported in previous RCTs of alkylat- intake 0.8–1.0 mg/kg per day‡
ing agents in membranous nephropathy 34,35. In 1996, the • Dual renin–angiotensin system blockade with
International Conference on Harmonization of Technical maximum tolerated doses of ACEIs and ARBs
Requirements for Registration of Pharmaceuticals for -- Start and up‑titrate an ACEI (or ARB)‡
-- Start and up‑titrate an ARB (or ACEI)‡
Human Use defined the Guidelines for Good Clinical
Practice, including monitoring of SAEs36. Before this • Start and up‑titrate other antihypertensive agents to
achieve the maximum tolerated reduction in blood
time, the under-reporting of such events could have
pressure (consider dihydropyridine calcium-channel
contributed to the general underestimation of the risks blockers as a last resort)
associated with heavy immunosuppression. Similarly,
• Add a lipid-lowering agent
in countries that formally adopted the guidelines in the
past few years37, more-recent (post‑1996) studies are *A response-driven, individually tailored approach to target
proteinuria <0.3 g per 24 h. ‡Before moving to the next step,
also likely to be affected by underestimation of adverse
check serum potassium levels and optimize metabolic acidosis
events38. Even in well conducted trials33, adverse events and hyperglycaemia control to minimize the risk of
such as lymphomas and other malignant diseases, which hyperkalaemia. ACEI, angiotensin-converting-enzyme
can occur as late as 10–20 years after treatment, are likely inhibitor; ARB, angiotensin-receptor blocker.
to be missed39. The risk of malignancies and other seri-
ous complications — including post-transplant lympho­
proliferative disorders, opportunistic infections, and function or severe, disabling symptoms of nephrotic
osteomuscular disease — increases when patients who syndrome. Over 10 years, the rate of remissions was
progress to ESRD receive a kidney transplant and receive similar in patients given immunosuppressive therapy
immunosuppressive therapy to prevent graft rejection. and in those who received conservative therapy alone,
Thus, the risk:benefit profile of combination ther- despite membranous nephropathy being less severe at
apy with steroids and chlorambucil is poor, particularly baseline in the conservative therapy group. However,
in patients at increased risk of treatment-related SAEs severe adverse events were remarkably more frequent
because of kidney function impairment or rapid renal in patients who underwent immunosuppression. In
function loss (the majority of patients enrolled in the UK particular, 15% of the 91 patients given cyclophospha-
trial33). Even in patients with a normal and stable glo- mide developed a malignancy compared with only 3%
merular filtration rate, the adverse effects of chlorambu- of the 130 who received conservative therapy. This dif-
cil have led the nephrology community to recommend ference was statistically significant, and in eight patients
that chlorambucil should not be used in the treatment the malignancy was fatal. Moreover, patients receiving
of membranous nephropathy 10. An adequately powered cyclophosphamide had higher rates of hospitalization,
RCT to assess the long-term benefits and risks of com- infection, bone marrow and liver toxicity, and cardio-
bination therapy with steroids and cyclophosphamide vascular and thrombotic events than patients who were
would require hundreds of patients to be followed‑up treated conservatively 45. Therefore, even when immu-
for 10–20 years40, but such a trial would not be justified nosuppression is restricted to patients at increased risk
given the availability of novel and safer treatments41. of disease progression, SAEs remain an important issue.
Indeed, since the late 1990s42, research has been tar- In this study 45, average patient exposure to cyclophos-
geted towards the identification of novel treatments phamide exceeded the cumulative exposure of the tra-
that achieve the same benefits as existing therapies ditional cyclical cyclophosphamide regimen10. However,
but with substantially fewer adverse effects. Improved the risk of SAEs remained when analyses were restricted
understanding of disease mechanisms43 has provided a to patients who received a cumulative dose of <20 g
strong rationale for more specific approaches that will cyclophosphamide or when the observation period was
potentially address important unmet needs in patients restricted to the first 3 months or 6 months of immuno-
with membranous nephropathy, particularly for those suppressive therapy 41. In this context, cyclophosphamide
with persistent nephrotic syndrome and worsening has been associated with an increased risk of malignancy
­kidney function44. even at cumulative doses of 12 g (REF. 46). Historical treat-
The Kidney Disease: Improving Global Outcomes ment approaches in membranous nephropathy have
(KDIGO) guidelines published in 2012 confirmed that been comprehensively reviewed elsewhere47,48.
treatment with alkylating agents should be restricted
to patients with persistent nephrotic syndrome10. The Towards disease-specific therapy
rationale is that, in these patients, the risks of treat- Membranous nephropathy was first described as a spe-
ment would be counterbalanced by the reduced risk cific disease entity in 1957 by David Jones49. In 1959,
of ESRD. In fact, a long-term, single centre, cohort the finding that rats injected with an extract of proxi-
study showed that a restrictive approach prevented mal tubular cells induced the deposition of subepithelial
51% of 254 patients with membranous nephropathy immune complexes that were similar to those observed
from receiving unneces­sary treatment 45. In this series, in humans with membranous nephropathy, strongly
steroids and cyclophosphamide were administered for suggested the possibility of an immune-mediated
6–12 months only to patients with worsening kidney patho­genesis of the disease50. These immunocomplexes,

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 565


©
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

Table 1 | Treatment of membranous nephropathy: conventional immunosuppressive protocols*


Regimen Drug Dose Schedule
Alkylating agents plus steroid combination therapy ‡

Chlorambucil Chlorambucil 0.2 mg/kg per day Months 2, 4, and 6


cyclical therapy
Prednisolone 0.5 mg/kg per day Months 1, 3, and 5
Methylprednisolone 1 g IV Three consecutive days at start
of months 1, 3, and 5
Cyclophosphamide Cyclophosphamide 2.5 mg/kg per day§ Months 2, 4, and 6
cyclical therapy
Prednisolone 0.5 mg/kg per day Months 1, 3, and 5
Methylprednisolone 1 g IV Three consecutive days at start
of months 1, 3, and 5
Cyclophosphamide Cyclophosphamide 1.5 mg/kg per day Months 1–6||
daily therapy
Prednisolone 0.5 mg/kg every second day Months 1–5, then taper dose
to stop in 6–8 weeks
Methylprednisolone 1 g IV Three consecutive days at start
of months 1, 3, and 5
Calcineurin inhibition therapy¶
Ciclosporin regimen Ciclosporin Initial dose 3.5 mg/kg per day, trough Months 1–6, then taper dose
level 125–225 μg/l by 25% each month; continue
treatment at 50% of dose until
12 months, then taper to lowest
possible maintenance dose#
Prednisolone** 0.15 mg/kg per day Months 1–6, then taper dose
(maximum of 15 mg)
Tacrolimus regimen Tacrolimus Initial dose 0.05 mg/kg per day, achieve Months 1–12, then taper to
trough level 3–5 ng/l; if remission is not lowest possible maintenance
achieved after 2 months, increase to dose||
5–8 ng/l
Prednisolone** 0.15 mg/kg per day Months 1–6, then taper dose
(maximum of 15 mg)
*Some immunosuppressive regimens require prophylactic measures to prevent adverse effects. ‡ Effectiveness documented in
controlled clinical trials, but with major and potentially fatal adverse effects; not recommended. §The Kidney Disease: Improving
Global Outcomes guidelines recommend 2 mg/kg per day10 .||Cyclophosphamide used for 12 months in REF. 45; treatment duration
is now limited to 6 months. ¶Unproven effectiveness with adverse effects, including nephrotoxicity; not recommended. #Incidence
of relapse is high; treatment must be continued in most patients. **Whether prednisolone coadministration is needed is unknown.
IV, intravenous.

however, contained IgG antibodies target­ing mega- Circulating autoantibodies against another human
lin, a protein expressed on both rat tubuli and podo- podocyte antigen — the thrombospondin type  1
cytes51, but not on human podocytes. A target antigen domain containing 7A protein (THSD7A) — have been
in humans was first identified in 2002 in the baby of subsequently observed in 5–10% of patients with mem-
a woman with a neutral endopeptidase (NEP) defi- branous nephropathy who do not have circulating anti-
ciency 52. The finding that anti-NEP alloantibodies pro- PLA2R autoantibodies56. The finding that most patients
duced by the mother crossed the placenta and bound to with membranous nephropathy have an autoimmune
NEP expressed on fetal podocytes52 confirmed the role response against either PLA2R or THSD7A, but very
of autoantibodies in the pathogenesis of membranous rarely against both57, suggests that both antigens can be
nephropathy in humans. the primary target of specific autoimmunity and con-
In 2009, circulating autoantibodies against the firms that PLA2R-associated membranous nephropathy
M-type phospholipase A2 receptor (PLA2R), primarily and THSD7A‑associated membranous nephropathy are
of the IgG4 subclass, were detected in ~70% of patients separate disease entities2.
with membranous nephropathy 43. Antibodies were dis- Several studies have since documented that anti-
ease specific as they could not be found in patients with PLA2R antibody titre correlates with disease activity
other proteinuric nephropathies43, but were associated and patient outcomes53,58–65. Low autoantibody levels at
with membranous nephropathy in several cohorts of diagnosis predict spontaneous remission66,67, whereas
patients of different ethnicities53,54. The pathogenic role high baseline anti-PLA2R antibody levels correlate with
of the PLA2R antigen was confirmed by the significant a reduced probability of spontaneous remission66, are
association found between single nucleotide poly­morph­ associated with progression to nephrotic syndrome
isms in the PLA2R gene and the development of the in patients with initial non-nephrotic proteinuria64,
disease55. and predict a high risk of relapse and progressive loss

566 | SEPTEMBER 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

Antibody-secreting CysR domain were younger, had less severe proteinuria, a


cell (including higher rate of spontaneous remissions, and a lower risk of
plasmablasts, plasma
Immature Mature Activated progression to ESRD than patients who also showed anti-
cells and long-lived
Pro-B cell Pre-B cell B cell B cell B cell body reactivity against CTLD1 and/or CTLD7 domains
memory cells)
CD20 CD38 indicated that the epitope profile could change over time
and that epitope spreading towards CTLD1 and CTLD7
might negatively affect disease outcome73. These findings
led to the hypothesis that antiPLA2R reactivity restricted
BAFF-R,
to the CysR domain might be associated with initial and
BCMA BLyS Anti-PLA2R milder stages of the disease, whereas additional immune
or TACI challenges might induce antigen intramolecular spread-
Anti-THSD7A ing towards the C‑terminus. Thus, as observed in other
human immune diseases74 and Heymann nephritis —
Targets for anti-BLyS Targets for anti-CD38 an experimental counterpart of human membranous
antibodies (e.g. belimumab) antibodies
nephropathy 75 — the emergence of new epitopes, such
(e.g. daratumumab
Targets for anti-CD20 monoclonal antibodies or isatuximab) and as CTLD1 and CTLD7 in patients with membranous
(e.g. rituximab or ofatumumab) protease inhibitors nephropathy might be associated with disease worsen-
(e.g. bortezomib) ing and decreased responsiveness to therapy 73. Thus,
qualitative evaluation of antiPLA2R reactivity through
Figure 1 | Targets for monoclonal antibodies in B-cell lineages. B cells emerge from
Nature
bone marrow stem cells as pro‑B cells and mature into pre‑B cells, ReviewsB cells,
immature | Nephrology
mature the development of epitope-specific anti-PLA2R assays,
B cells, and activated B cells. Eventually, they develop into antibody-secreting cells, in combination with the measurement of overall anti-
including plasmablasts, plasma cells, and long-lived memory plasma cells that secrete IgA, PLA2R titre, might improve monitoring of disease activity
IgE, IgG, and IgM antibodies. Autoreactive B‑cell clones can produce anti-M-type and response to therapy. Whether similar considerations
phospholipase A2 receptor (PLA2R) and anti‑thrombospondin type 1 domain containing 7A might apply also to the THSD7A antigen is unknown.
protein (THSD7A) antibodies and conceivably, autoantibodies against unknown antigens In a long-term cohort study of 132 patients with
expressed on podocytes. As B cells mature, they develop various markers on the cell surface membranous nephropathy and chronic nephrotic
that can become targets for specific monoclonal antibodies. Anti‑CD20 monoclonal syndrome, a lower antibody titre strongly predicted a
antibodies, such as rituximab and ofatumumab, bind and kill CD20‑expressing B cells higher rate of complete or partial remission and a faster
(pre‑B cells, and immature, mature, and activated B cells), but not plasmablasts, mature
progression to the event following treatment with the
plasma cells or memory plasma cells as they do not express this antigen. Plasma cells
express CD38 and might, therefore, be a target for anti‑CD38 antibodies such as anti‑CD20 monoclonal antibody rituximab65. Moreover,
daratumumab and isatuximab. Immature, mature, and activated B cells express receptors the reduction or depletion of anti-PLA2R autoantibodies
for the B lymphocyte stimulator (BLyS, also known as BAFF) such as B cell activating factor at 6 months strongly predicted disease remission during
receptor (BAFF‑R), B cell maturation antigen (BCMA), and the transmembrane activator follow‑up, and a 50% decrease in antibody titre preceded
and calcium-modulating cyclophilin ligand interactor (TACI). The anti-BLyS monoclonal an equivalent decrease in proteinuria by 10 months65.
antibody belimumab might, therefore, prevent B-cell differentiation into plasma cells by Sensitivity analyses confirmed that short-term changes
blocking the interaction between this lymphocyte stimulator and its receptors. Proteasome in antibody titre strongly predicted long-term disease
inhibitors, such as bortezomib, can prevent antibody production by inducing plasma cell outcome (complete remission was considered a single
apoptosis, whereas anti‑CD38 monoclonal antibodies, such as daratumumab and end point, and remission of nephrotic syndrome com-
isatuximab, can directly induce plasma cell cytolysis.
bined with an increase in serum albumin to normal
levels was a more restrictive end point in patients with
hypoalbuminaemia at baseline)65. Response to treatment
of kidney function63,64,68. Moreover, decreasing anti- was similar in patients with or without detectable anti-
PLA2R antibody levels strongly predict remission of bodies or for whom antibody data were not available65.
protein­uria53,62,69 and response to various traditional These findings strongly suggest that in a substantial pro-
and novel CD20‑targeted immunosuppressive treat- portion of patients who are anti-PLA2R negative, mem-
ments62,65. Preliminary data suggest the same will be true branous nephropathy is sustained by other, unknown
for anti‑THSD7A antibody levels in predicting disease autoantibodies, and that the production of these anti-
activity and response to therapy 56,70. bodies is affected by rituximab therapy to a similar
Disease severity and response to treatment of patients extent as is anti-PLA2R.
with membranous nephropathy is influenced not only by After almost 40 years of empirical treatment, the dis-
the overall titre of circulating anti-PLA2R autoantibodies covery of anti-PLA2R and anti‑THSD7A autoanti­bodies
but also by the nature of these antibodies. PLA2R1 is a provided the first clear pathophysiological rationale for
membrane receptor that has a large extracellular region interventions specifically aimed at preventing anti-
which includes a cystein-rich domain (CysR) and eight body production (FIG. 1) and subepithelial deposition of
different C‑type lectin domains (CTLD1–8)71. The CysR antibody–­antigen immunocomplexes (FIG. 2). Such treat-
domain is probably the dominant epitope of the PLA2R1 ments could potentially prevent complement activation
antigen72; However, site-directed mutagenesis experi- and podocyte damage, eliminate nephrotic syndrome,
ments have identified CTLD1 and CTLD7 as additional and even resolve the glomerular pathology of mem-
and distinct PLA2R1 epitopes that could act as potential branous nephropathy 76,77. In this context, combined
targets of anti-PLA2R autoreactivity73. The finding that assessment of circulating anti-PLA2R or anti‑THSD7A
patients with isolated anti-PLA2R reactivity against the autoantibodies and proteinuria, and serum albumin level

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 567


©
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

a b
Podocyte
Immuno-
complexes

8
7 8
6 7
5
4 5
6
8 Complement
activation
3 4 7
2 3 6
1 2 5
1 4
3
2
1

Podocyte
damage

Proteinuria

Anti-PLA2R against PLA2R CysR domain PLA2R CysR domain PLA2R FNII domain

Anti-PLA2R against PLA2R CTLD1 domain PLA2R CTLD domains (1–8)

Anti-PLA2R against PLA2R CTLD7 domain PLA2R transmembrane and intracellular domains

Figure 2 | Prevention of antigen–antibody immunocomplex deposition on the glomerular basement membrane.


Nature Reviews | Nephrology
a | A glomerular capillary and its basement membrane surrounded by podocytes. Anti-M-type phospholipase A2
receptor (PLA2R) antibodies are shown in the capillary lumen. b | Binding of circulating anti PLA2R autoantibodies to their
specific cystein-rich domain (CysR), C-type lectin domain (CTLD)1 or CTLD7 epitopes expressed by the PLA2R receptor
on the podocyte cell surface leads to the deposition of antigen–antibody subepithelial immunocomplexes that activate
the complement system with secondary podocyte damage and proteinuria. In theory, antibody–antigen binding and the
deposition of subepithelial immunocomplexes could be prevented by epitope-specific ligands that bind circulating
antiPLA2R antibodies and prevent their binding to epitopes of the PLA2R antigen.

in patients with hypoalbuminaemia, could be instru- consistently shown that antibodies produced by auto­
mental in monitoring disease activity and guiding per- reactive B-cell clones initiate events resulting in injury to
sonalized therapy with conventional immuno­suppressive the glomerular barrier and the development of protein­
protocols or specific B cell-targeting monoclonal anti- uria79. Consistent with these observations, cyclophos-
bodies78. Indeed, antibody levels provide a prompt and phamide has a direct inhibitory effect on B cell antibody
reliable indication of immunological status and an inte- production in membranous nephropathy, adding to
grated evaluation of serology and proteinuria might the non­specific antimitotic and immunosuppressive
accelerate therapeutic decision-making and address properties that mediate many of the adverse effects of
diagnostic uncertainties when proteinuria and antibody this drug 80. The availability of a monoclonal antibody
levels are discordant. Specifically, persistent proteinuria against the B-cell surface antigen CD20 (REF. 81) has ena-
in the face of low antibody levels might reflect chronic bled investigation into whether targeted B‑cell depletion
damage, whereas high antibody levels in the absence of with inhibition of nephritogenic autoantibody produc-
clinical symptoms could signal an impending relapse. tion improves the outcome of patients with membran­
This integrated approach to membranous nephropathy ous nephropathy while avoiding the adverse effects of
could increase diagnostic and prognostic accuracy, limit ­steroids and immunosuppressants (TABLE 2).
unnecessary exposure to immunosuppressive therapy, Against this background, the anti‑CD20 mono­clonal
optimize efficacy of treatment, and minimize the risk or antibody rituximab was tested in eight patients with
severity of recurrent disease in allotransplants78. These membranous nephropathy and long-term proteinuria42.
theories can now be addressed in prospective studies Over 20 weeks of follow‑up, proteinuria decreased by 62%
designed to test whether treatment titration to auto­ and serum albumin level increased by 31% compared to
antibody titre will increase the probability of achieving baseline levels. In two patients proteinuria decreased
remission from nephrotic syndrome and prevent relapse to <1 g per 24 h and in three patients to <3.5 g per 24 h,
while decreasing the risk of treatment-related adverse and no SAEs occurred42. The reduction in proteinuria
effects. In antibody-negative patients, proteinuria and was sustained over 1 year and was associated with stable
serum albumin levels remain the most sensitive markers kidney function and a reduction in body weight, blood
of disease severity and outcome. pressure, and serum cholesterol level82. These seminal
observations were confirmed by subsequent studies22,23,83.
Monoclonal antibodies: rituximab Among 35 patients with membranous nephropathy (half
Prior to the finding that anti-PLA2R autoantibodies of whom had not been responsive to immunosuppres-
have a central role in the pathogenesis of human mem- sant therapy), rituximab administered at a dose of two
branous nephropathy 43, data from animal studies had 1 g infusions 2 weeks apart, or four once-weekly infusions

568 | SEPTEMBER 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

Table 2 | Treatment for membranous nephropathy: B cell and plasma cell-targeted protocols
Target Drug Dose Schedule
Anti‑CD20 monoclonal Rituximab • 375 mg/m intravenously
2
• Every week for 4 weeks
antibodies* • 375 mg/m2 intravenously • B cell-driven treatment‡
• 1,000 mg intravenously • Days 1 and 15
Ofatumumab 300 mg intravenously B cell-driven treatment‡
Anti-BLyS monoclonal Belimumab 10 mg/kg intravenously • Every 4 weeks (if urinary
antibodies§ protein:creatinine ratio <1,000
mg/mmol)
• Every 2 weeks (if urinary
protein:creatinine ratio >1,000
mg/mmol)
Plasma cell-targeted protocols Bortezomib 1.3 mg/m2 subcutaneously Four doses over 2 weeks
(proteasome inhibitors||)
*Safe and well tolerated. Effectiveness of rituximab proven in randomized trials and observational studies. Preliminary observational
evidence of efficacy for ofatumumab in rituximab-resistant forms of membranous nephropathy. ‡A second 375 mg/m2 dose of
rituximab or a second dose of 300 mg of ofatumumab are administered 1 week after the first infusion if >5 circulating B cells per mm3
remain. §Preliminary evidence of efficacy from a small, randomized pilot study; no long-term data available. ||Preliminary observational
evidence of efficacy in rituximab-resistant forms of membranous nephropathy; serious adverse effects. BLyS, B lymphocyte stimulator.

of 375 mg/m2, achieved complete or partial remission of Irrespective of the mechanisms of drug action, a
proteinuria in 50% and 80% of patients at 1 year and at matched cohort study showed that rituximab safely and
2 years, respectively 22,23. The reduction in proteinuria was persistently reduced proteinuria in untreated patients and
similar with the two dosing regimens, and was gradual in those who did not respond to immunosuppressive ther-
and sustained over time. The recovery of B-cell counts apy or relapsed after transient remission88. Among 100
once treatment was completed, however, was faster in patients with membranous nephropathy and nephrotic
patients with proteinuric membranous nephropathy than syndrome, the rates of remission were similar with ritux-
in patients without proteinuria treated with rituximab, imab as a first-line therapy and as a ‘rescue’ therapy 86.
such as those with rheumatoid arthritis, non-Hodgkin Rituximab also enabled successful withdrawal of tacroli-
lymphoma, or antineutrophil cytoplasmic antibody mus or ciclosporin in 13 calcineurin inhibitor-dependent
(ANCA)- associated vasculitis22,23,84,85. This finding sug- patients89. Three patients experienced relapse of nephrotic
gests that rituximab is lost in the urine of patients with syndrome over a period of 35 months but fully recovered
severe proteinuria. However, rituximab levels did not following a second course of rituximab89.
correlate with treatment response22,23. Together, these data indicate that selective B‑cell
In a single-centre cohort study published in 2012, depletion by rituximab is at least as effective as other
B‑cell depletion led to complete or partial remission conventional immunosuppressive regimens in promot-
of nephrotic syndrome in 65% of 100 patients, and an ing disease remission in patients with membranous
additional 20 patients experienced a >50% reduction nephropathy and severe nephrotic syndrome, even
in proteinuria86. The median duration of proteinuria after other treatments have failed. Importantly, clinical
before rituximab administration was >2 years (~6 years benefits are associated with regression of the histologi-
in those previously exposed to other immunosuppressive cal lesions characteristic of membranous nephropathy,
regimens), and all patients had been taking ACE inhibi- along with decreased glomerular IgG4 and C3 staining,
tors or ARBs for at least 6 months. Therefore, remission reabsorption of electron-dense subepithelial deposits, an
of nephrotic syndrome in these patients was unlikely increased number of podocyte slit diaphragms, and an
to have been spontaneous. Again, the antiproteinuric increased percentage of electron-dense slit diaphragms76.
effect was progressive over time and, at the 4‑year follow These findings, combined with data showing that a
up, all patients had achieved complete or partial remis- decrease in albumin fractional clearance is correlated
sion86. Data from other studies indicating that depletion with an increase in electron-dense diaphragms, pro-
of anti-PLA2R antibodies precedes remission of the vides evidence that rituximab interferes with the mech-
nephrotic syndrome59,65 and that their re-emergence anisms of membranous nephropathy 76. Regression of
precedes disease recurrence65, provides convincing evi- histological changes was also observed in isolated cases
dence of a causal relationship between anti-PLA2R anti- of membranous nephropathy described decades before
body production and disease activity. The finding that rituximab became available for clinical use90–92; how-
the reduction in proteinuria up to the point of complete ever, whether they reflect treatment-induced or spon-
remission follows B‑cell depletion by several months is taneous remission of the disease is difficult to establish.
consistent with evidence that the resolution of in situ By contrast, although ciclosporin was shown to effec-
immune deposits is a slow and progressive phenom­enon tively reduce proteinuria in patients with membranous
that occurs several months after transplantation of kid- nephropathy, repeat renal biopsy demonstrated that
neys from rats with experimental membranous nephro­ electron-dense immune deposits associated with the glo-
pathy (Heymann nephritis) into syngeneic rats without merular capillary wall were more numerous and larger
the disease87. after ciclosporin therapy than before93.

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 569


©
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

An important, but frequently neglected benefit of 10 mg of chlorphenamine and 500 mg of hydrocortisone
rituximab is its major impact on patient quality of life94. minimizes the risk of these events. SAEs occurring after
Patients receiving rituximab benefit from the improve- exposure to rituximab are rare. In 100 patients treated
ment in symptoms associated with remission of protein- with rituximab77, the percentage who experienced an
uria and discontinuation of other immunosuppressive SAE was lower than in the supportive therapy group of a
medications. By contrast, in patients treated with ster- different trial33 (11% versus 42% over 29 and 36 months,
oids, alkylating agents or calcineurin inhibitors, quality respectively). Moreover, the finding that SAEs clustered
of life is detrimentally affected by the toxicities of these in the subgroup of patients who did not achieve remis-
medications, even when remission is achieved94. sion strongly suggests that these events were related to
the underlying disease rather than to treatment. The
Comparative safety of rituximab rate of infections and cardiovascular events was actually
Rituximab is generally well tolerated. In five randomized reduced in patients who achieved remission86.
controlled trials95–99, the average incidence of severe The risk of SAEs such as malignancies that occur
infections in patients treated with rituximab (10%) as late as 10–20 years after treatment 39 and, therefore,
was comparable to that observed in patients receiving are not captured during the observation period, can-
placebo (12%). A study of case descriptions of patients not be excluded from trials of steroids and alkylating
treated with rituximab from 1997 to 2008 identified 57 agents32,35,38. However, data from patients receiving life-
patients with progressive multifocal leukoencephalopa- long rituximab therapy for chronic lymphomas, or those
thy (PML)100. 52 of these patients had lymphoproliferative exposed to high cumulative doses for the treatment of
disorders and five had autoimmune diseases. All of the autoimmune diseases or lymphoproliferative disorders
patients had previously been exposed to corticosteroids over a >10‑year period, show that rituximab is safer than
and several chemotherapeutic agents, including cyclo- other immunosuppressants107–109. In a large cohort of
phosphamide in 74% and chlorambucil in 21% of cases, patients with rheumatoid arthritis who were followed for
in various combinations. No patient received rituximab 11 years, rituximab was not associated with an increased
as the only immunosuppressive medication. Moreover, risk of cancer 110. In another study, three malignancies
it is unknown how many patients —conceivably sev- observed over a median of 29 months in 100 consecu-
eral thousands — had been exposed to rituximab at the tive patients with membranous nephropathy treated with
same centres over the observation period without devel- rituximab86 is likely to reflect the age-adjusted incidence
oping PML. On the basis of this observational study, it of cancer in the general population111. Of note, the patient
is impossible to establish a causal relationship between who eventually died of lung cancer had been previously
exposure to rituximab and development of PML100. On treated with steroids and alkylating agents. By contrast,
the other hand, >30 cases of PML have been described combined therapy with steroids and cyclophosphamide
in patients with vasculitis associated with systemic lupus in membranous nephropathy confers a threefold increase
erythematosus (SLE) or ANCA who were treated with in the risk of cancer compared with that of the general
cyclophosphamide-based regimens101,102. In the case of a population, which translates to an annual increase in
child with steroid-dependent idiopathic nephrotic syn- risk of 0.3–1.0%46. This finding is particularly relevant
drome, exposure to other immunosuppressant drugs is for patients with membranous nephropathy who relapse
likely to have been involved in the development of fulmi- after treatment with cyclophosphamide, because sub-
nant enterovirus myocarditis requiring heart transplan- sequent treatment will result in a cumulative dose of
tation 13 months after rituximab therapy 103. In fact, the ­cyclophosphamide well above the safety threshold.
finding that replication of John Cunningham virus and Within the limitations of comparisons across studies,
BK virus is not affected by rituximab therapy in children rituximab seems to be at least as effective as conventional
with recurrent idiopathic nephrotic syndrome104 indi- treatments for membranous nephropathy, but is safer.
cates that complications related to viral reactivations Severe complications, such as PML, pneumocystis pneu-
should not be more frequent with rituximab than with monia, and pulmonary fibrosis that have been ascribed
conventional immunosuppressants. In an RCT of chil- to rituximab112 were observed in patients with lympho­
dren with frequently relapsing nephrotic syndrome, no proliferative113 or autoimmune diseases114, or those who
significant increase in the incidence of severe adverse had undergone bone marrow transplantation115, all of
events was found with rituximab compared with pla- whom had been exposed to other immunosuppressant
cebo105. The incidence of infections was also comparable drugs. No such events have been reported in patients
between the two groups105. Moreover, the risk of infec- receiving rituximab monotherapy, including those with
tions and other complications is reduced when rituxi- membranous nephropathy. Anecdotal evidence sug-
mab is used to aid ­withdrawal from steroids and other gests that, by allowing withdrawal of steroid and other
immunosuppressants106. immuno­suppressants, rituximab may even favour remis-
In the context of membranous nephropathy, the sion of some of the above complications106, including
only adverse effects that can be ascribed with certainty PML (P. Ruggenenti and G. Remuzzi; unpublished work).
to rituximab are infusion-related events such as hypo-
tension, cutaneous rash, and bronchial wheezing 86. Optimal dosing of rituximab
These effects are not serious and resolve with tempo- Rituximab treatment protocols for patients with mem-
rary interruption of the infusion or, in exceptional cir- branous nephropathy vary widely. The most frequently
cumstances, with hydrocortisone86. Premedication with used protocol involves a dose of 375 mg/m2 once a

570 | SEPTEMBER 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

week for 4 weeks, but eight-dose ‘prolonged protocols’, Controlled studies of rituximab
or ‘extended protocols’ with monthly infusions for 2 or The GEMRITUX trial. In the prospective, randomized,
3 months following the standard four-dose regimen controlled GEMRITUX clinical trial25, the effects of two
have also been used116. However, CD20+ cells were found 375 mg/m2 doses of rituximab were compared to non­
to be absent from the circulation after a single dose of immunosuppressive antiproteinuric treatment (NIAT)
rituximab in patients with membranous nephropathy on the rate of complete or partial remissions at 6 months
or SLE117,118. This finding questions the need for addi- (primary end point) in 75 patients with membranous
tional doses of rituximab, which could increase the risk nephropathy and residual proteinuria (>3.5 g per 24 h)
of adverse effects or sensitization and treatment costs, despite maximal tolerated doses of ACE inhibitors or
without conferring additional benefits. ARBs, statins, and diuretics. The treatment effect was
To address this question, a prospective 1:2 evaluated after this short period of follow up so that
matched-cohort study compared the risk:benefit profile patients with persistent nephrotic syndrome could be
of a B cell-driven rituximab treatment with the standard offered rescue treatment with other immunosuppressive
four 375 mg/m2 dose rituximab protocol in 36 patients medications (particularly those in the control group who
with membranous nephropathy and nephrotic syndrome received supportive NIAT only). At 6 months, numerically
refractory to conventional therapy 119. Patients allocated more patients in the rituximab group than in the control
to the B cell-driven protocol received a second infusion group reached the primary end point (13 versus eight), but
of rituximab only if they had more than five B cells per this difference was not significant25. By 17 months, how-
mm3 of peripheral blood after the first 375 mg/m2 dose, ever, the rate of complete or partial remission in rituxi-
which occurred in only one of the 12 patients in this mab group was almost twice that of controls (64.9% versus
group119. The two treatments promptly and persistently 34.2%), and the difference in remission rate was significant
depleted circulating B cells in all patients and achieved (P = 0.03). Seven patients treated with rituximab achieved
a similar time-dependent reduction in proteinuria. complete remission compared to just one control25. This
However, adverse events and hospitalizations were less finding was not surprising, because the antiproteinuric
frequent with the B cell-driven approach, which was also effect of rituximab can take at least 12 months to fully
fourfold less expensive119. Because of the remarkable manifest86, an effect that is largely preceded by depletion
reduction in treatment costs (~€10,000 or ~US$13,000 of circulating anti-PLA2R antibodies and an increase in
per patient 119), the B cell-driven protocol should facil- serum albumin concentration65. The time lag between the
itate access to rituximab even in resource-limited set- start of immunosuppression and reduction in protein­
tings. Indeed, one 375 mg/m2 dose of rituximab in an uria has been demonstrated in every study conducted in
average 70 kg patient costs €3,206 ($4,275)120. The alter- patients with membranous nephropathy 22,32,86,125. In each
native treatment with intravenous and oral steroids plus of these studies, the nadir of proteinuria in patients who
oral cyclophosphamide for 6 months at currently rec- responded to immunosuppression was not reached until
ommended doses costs approximately €450 ($600)121. at least 18 months after the start of treatment, regardless
However, this figure does not include the cost of fre- of the type of drug. Therefore, studies of membranous
quent clinic visits to monitor blood cell counts and hos- nephropathy in which the primary outcome measure is
pitalizations because of treatment-related adverse events. remission of proteinuria should not have an end point
Considering that a 1‑day hospitalization in a noninten- earlier than 18 months from the start of therapy. These
sive care unit, including a nephrology and dialysis unit, considerations explain why the GEMRITUX trial38 could
costs €300–500 ($400–666)122, just one admission would not have achieved its primary end point.
largely offset the costs saved with steroids plus alkylating In the rituximab group, anti-PLA2R autoantibodies
agents compared to the cost of rituximab therapy. were almost fully depleted from the circulation 3 months
The current recommendation for patients with after randomization, whereas the antibody titre did not
membranous nephropathy is to use calcineurin change appreciably in the control group throughout the
inhibitors for at least 12 months before considering observation period (FIG. 3). Serum albumin concentration
discontinuation10. In the USA, the average cost of a increased in rituximab-treated patients at 3 months and
calcineurin inhibitor at doses used for membranous 6 months compared with baseline, but did not change
nephropathy is ~$700 per month123. To this expense in control patients. As previously observed with ACE
must be added the cost of monitoring drug levels and inhibitors24, the increase in serum albumin preceded
of blood pressure-lowering medications needed to con- the reduction in proteinuria by months25. A post-hoc
trol treatment-induced hypertension. In the long-term, analysis demonstrated that at 6 months, more patients
treatment costs will increase as >50% of these patients receiving rituximab than in the supportive therapy
relapse within 12 months and 18 months after with- group achieved a >50% reduction in proteinuria with
drawal of ciclosporin and tacrolimus, respectively 124,125. at least a 30% increase in serum albumin levels (40.5%
These relapses require further courses of calcineurin versus 13.2%; P <0.01)25.
inhibitors and multiple relapses are associated with a Notably, the rate and timing of complete or partial
doubling of serum creatinine level and progression to remission achieved by rituximab was similar to that pre-
ESRD48,126. Thus, additional costs will be incurred with viously reported with rituximab in uncontrolled series127
the need for repeated courses of calcineurin inhibitors and also to those reported in previous trials with ster-
and to manage complications related to the chronic oid and alkylating agents at 6 months and 1 year of fol-
nephrotoxicity of these agents. low up32,38. Remarkably, the safety profile of rituximab

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 571


©
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

a b St‑Cp group than in the rituximab group (46 versus 11


P = 0.03
20 70
events, P <0.001). Moreover, 83% of the SAEs observed in
(percent change versus baseline)
Serum anti-PLA2R antibody level

Patients achieving complete or


partial remission of nephrotic
Month 3 (n = 48) Month 6 (n = 51)
0 the St‑Cp group were likely or possibly treatment-related,
60
NIAT and rituximab whereas all the SAEs observed in the rituximab group

NIAT

syndrome (%)
–20 were likely unrelated to treatment. In particular, six cases
50
–40
of cyclophosphamide-related myelotoxicity occurred in
40
addition to ten cases of hyperglycaemia, seven throm-
–60 boembolic events and one case of osteonecrosis, which
–80 30 were conceivably related to concomitant steroid ther-
apy 41. Consistent with the findings of the GEMRITUX
–100 P<0.001 P<0.01 20 trial25, the rate of infections in the rituximab group was
Figure 3 | Key findings from the GEMRITUX trial . a | Changes in serum anti-M-type
38 similar to that in the general population, and these events
phospholipase A2 receptor (PLA2R) antibody levels at 3 months Nature Reviews | after
and 6 months Nephrology were unlikely to be related to treatment, whereas infec-
patients were randomly assigned to receive nonimmunosuppressive antiproteinuric tions in steroid and cyclophosphamide-treated patients
therapy (NIAT) or NIAT plus rituximab. b | Percentage of patients achieving complete or were more likely to be severe and to result in hospital
partial remission of the nephrotic syndrome at the end of the extension period admission (11serious infections, including three fatal
(17 months). In the rituximab group, anti-PLA2R autoantibodies were almost fully depleted cases of sepsis, in addition to 37 nonserious infections).
from the circulation 3 months after randomization. The treatment effect was sustained up Moreover, three blood malignancies and five solid
to 6 months, whereas the antibody titre did not change appreciably in the control group tumours (fatal in two patients) were observed in the
throughout the observation period. By 17 months after randomization the rate of
steroid and cyclophosphamide group. All these cancers
complete or partial remission in the rituximab group was almost twice that of controls
and the between-group difference in remission rate was significant.
were thought possibly related to treatment. One cancer
was thought likely to be related to previous chlorambucil
exposure. Taken together, these findings support the well-­
therapy was similar to that of supportive therapy, a find- established carcinogenic effects of alkylating agents46,128.
ing that supports safety data from previous uncontrolled In this study, cumulative patient exposure to steroids and
studies22,23,42,65,82,83,86. cyclophosphamide exceeded the limit recommended in
the KDIGO guidelines10. According to these guidelines,
Comparative analyses with steroid and cyclophospha‑ a 75 kg patient treated with 2 mg/kg per day of cyclophos-
mide. To date, the safety and efficacy profile of rituximab phamide for 6 months would be exposed to a cumulative
compared to that of conventional therapy (steroids and dose of 27.4 g. Patients in this study received 1.5 mg/kg per
alkylating agents) has not been compared in an RCT. day of cyclophosphamide and were exposed to a cumu-
Therefore, data have been compared from two well-­ lative dose ranging from 10.2 g at 3 months to 20.5 g at
defined cohorts of patients with membranous nephro­ 6 months and, ultimately, 41.1 g at 12 months. Thus,
pathy and persistent nephrotic syndrome, 100 of whom at 6 months the administered cumulative dose of cyclo-
were treated with rituximab and 103 with a steroid plus phosphamide was within recommended range10. Higher
cyclophosphamide (St‑Cp) combination regimen, and than recommended cumulative doses might result in an
who were monitored for up to 5 years according to pre- increase in long-term adverse effects. However, in this
defined standardized protocols45,86 at two different neph- comparative analysis most adverse events (such as leu-
rology units in Europe41. Cox regression analyses adjusted kopaenia, infections, and liver toxicity) occurred within
for baseline patient characteristics showed a marked dif- 3–6 months, the period of time when patient exposure
ference in safety profiles between the two groups. Over to cyclophosphamide was within recommended limits10.
a median follow‑up of 40 months, patients in the rituxi­ Moreover, no clear dose–response relationship was found
mab group had significantly fewer adverse events than the between cyclophosphamide and malignancy in the cumu-
St‑Cp group (63 versus 173; P <0.001), with differences in lative dose range between 10 g and 40 g (REF. 46). Therefore,
terms of both SAEs (11 versus 46; P <0.001) and nonseri- the excess long-term risk of malignancies associated with
ous adverse events (52 versus 127; P <0.001). The cumula- cyclophosphamide therapy could not be fully explained
tive incidence of any first (35.5% versus 69.0%; P <0.001), by patient exposure to high cumulative doses of the drug.
serious (16.4% versus 30.2%; P <0.002), or nonserious Notably, the adverse events associated with rituximab
(23.6% versus 60.8%; P <0.001) adverse event was sig- in studies reported since 2002 (REF. 42) are less frequent
nificantly lower with rituximab. Adjusted hazard ratios and less severe than those observed with combination
between rituximab and St‑Cp groups were 0.27 (95% CI steroid and cyclophosphamide therapy, and even with
0.16–0.44) for any first adverse event, 0.32 (95% CI 0.15– ciclosporin, in trials conducted before publication of the
0.68) for SAEs, and 0.23 (95% CI 0.13– 0.41) for nonseri- Guideline for Good Clinical Practice in 1996 (REF. 36).
ous adverse events. Although the cumulative incidence of Rituximab has been shown to be remarkably safer
partial remission was lower in the rituximab group, rates than ‘traditional’ medications used for decades for the
of complete remission and the composite renal end point treatment of membranous nephropathy, despite prob-
(doubling of serum creatinine compared to baseline levels, able under-reporting of their adverse effects in non-­
development of ESRD, need for chronic renal replacement monitored trials prior to the early 1990s. Rituximab
therapy or death from any cause) did not differ signifi- might, therefore, be considered the first-line therapy for
cantly between groups. In particular, the rate of SAEs and patients with membranous nephropathy and persistent
fatal adverse events was more than fourfold higher in the nephrotic syndrome despite conservative treatment.

572 | SEPTEMBER 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

Ongoing trials. The RI‑CYCLO trial will assess whether remission required retreatment. No serious infections
rituximab is safer and achieves complete remission of were associated with rituximab, and only nonserious
nephrotic syndrome more effectively than cyclical ster- events were observed early after treatment 140. This find-
oid and cyclophosphamide combination therapy at ing is remarkable considering that patients were already
1 year in 70 patients with membranous nephropathy 129. receiving triple antirejection therapy with corticoster-
MENTOR130 and the STARMEN trial131 will assess, oids, calcineurin inhibitors (or sirolimus in three cases),
respectively, whether rituximab is noninferior to ciclo- and mycophenolate mofetil with the exception of four
sporin in inducing long-term remission (complete or patients who were on a steroid-free regimen. The highest
partial) of proteinuria132 and whether, combined with risk of recurrence was observed in patients with high
tacrolimus, rituximab increases the rate of complete levels of proteinuria and, as previously reported141,142,
or partial remissions and improves survival with fewer detectable circulating anti-PLA2R antibodies at the time
adverse effects compared to steroids plus cyclophos- of transplantation. However, one-third of patients with-
phamide133. Of note, the patients enrolled in MENTOR out detectable anti-PLA2R antibodies also had disease
have better renal function than patients in the above- recurrence. In some patients, anti-PLA2R antibodies
mentioned trial from the UK33, and ciclosporin will be could not be detected after transplantation despite per-
administered at lower doses, which should reduce the sistent glomerular PLA2R antigen staining — probably
risk of chronic nephrotoxicity. because transplantation-related immunosuppression can
inhibit autoantibody production141 and potentially even
Rituximab and kidney transplantation improve the response to rituximab23,136.
Membranous nephropathy recurs in approximately 42% On the basis of these findings, patients with mem-
of patients who undergo kidney transplantation and branous nephropathy who are candidates for kidney
can lead to proteinuria, reduced graft function, and an transplantation should be advised that the cumulative
increased risk of graft failure134–138. Rituximab has been risk of disease recurrence is 50–60% over ~6 years, with
used successfully in these patients136,139,140. 30–35% of recurrences observed during the first year
Study of post-transplantation serial, per-protocol post-­transplantation140. Although 60% of membranous
biopsies has led a new understanding of subclinical nephropathy recurrences are progressive, in most cases
disease and histology at early stages of membranous rituximab is effective particularly when applied early in
nephropathy 139 and, importantly, has shown that rituxi­ the disease140. Close surveillance of proteinuria, albu-
mab therapy can result in both clinical remission and minuria, and antibody titre is advised, particularly in
histological resolution of disease136. Approximately patients at high risk of recurrence. Theoretically, ritux-
80% of patients with membranous nephropathy under- imab given before transplantation could prevent mem-
going kidney transplantation have anti-PLA2R anti- branous nephropathy recurrence and allograft injury 140.
bodies, which seem to predict disease recurrence and If this approach is considered, rituximab should be
response to anti‑CD20 therapy 59,141,142. Among 63 kid- administered several months before transplantation
ney graft recipients with biopsy-proven membranous because the response to therapy is progressive over time65.
nephropathy who were observed for up to 10 years, Taken together, the available data indicate that an
30 (46%) experienced a relapse that was confirmed by approach focused on early diagnosis and early B-cell
early per-protocol biopsy in 16 patients140. This obser- targeting therapy could substantially reduce the risk
vational study showed that per-protocol biopsy enables of kidney graft loss due to membranous nephro­pathy
early diagnosis, before progression to severe disease. recurrence. In the pre-rituximab era, this risk was
Disease recurrence often occurred during the first year reported to account for 18–20% of allograft losses143,144.
after transplantation, although a second ‘wave’ of recur-
rences were detected by per-protocol biopsies taken at Failure of rituximab therapy
5 years140. This observation might relate to the fact that When CD20‑targeted therapy with rituximab fails (in
induction immunosuppression can result in a reduction ~25–30% of patients with membranous nephropathy
in anti-PLA2R antibodies early after transplantation141. and nephrotic syndrome), the nephrologist is faced with
After recurrence, proteinuria did not progress in 29% of the dilemma of exposing the patient to toxic medica-
24 patients, which is similar to the rate of spontaneous tions or observing the patient progress while receiving
clinical resolution of membranous nephropathy reported conservative therapy. This issue is not trivial because
in native kidneys7. Approximately 70% of patients with nephrotic syndrome substantially reduces quality of life
recurrence of membranous nephropathy had either (particularly physical functioning) and is associated with
high-grade proteinuria or increasing urine protein serious cardiovascular and thromboembolic complica-
­levels over time. Based on the assumption that patients tions. Steroids plus cyclophosphamide and calcineurin
with proteinuria >1,000 mg per day have progressive inhibitors should be avoided owing to their associated
disease135,136, those who reached this level of proteinuria adverse effects. Moreover, no data are available on their
were treated with rituximab140. Treatment achieved dis- efficacy as second-line therapy for patients in whom
ease remission and histological resolution in 82% and rituximab treatment fails. If the patient is a potential
40% of patients, respectively, and prevented kidney graft candidate for kidney transplantation, conservative ther-
failure in all patients140, a remarkable change from previ- apy can be instrumental in avoiding the risk of add‑on,
ous experience134–138. Furthermore, the response to ther- adverse interactions between immunosuppression and
apy was persistent; no patient who achieved complete subsequent antirejection treatment.

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 573


©
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

Rituximab differentiate into plasmablasts (immature plasma cells),


binding site which invade the peripheral circulation, produce anti-
bodies, and further differentiate into mature, short-lived,
Ofatumumab
binding site nonproliferating plasma cells. Activated B-cell clones,
Large loop however, can be disrupted by nonspecific immuno­
B-cell suppressants, such as steroids and alkylating agents, as
membrane Small loop well as by anti‑B-cell antibodies, such as rituximab and
ofatumumab, which have a direct cytolytic effect. Other
antibodies, such as belimumab, can block growth factors
and mediators of B‑cell proliferation and differentiation
(discussed in further detail below). After disruption or
Intracellular inhibition of the autoreactive clones, the plasma cell pool is
no longer fueled; short-lived plasma cells disappear within
a few days of treatment and are no longer replenished.
1 297
By contrast, memory plasma cells are resistant to
N-terminal C-terminal conventional immunosuppression and B‑cell targeted
intervention. These cells secrete antibodies independent
Figure 4 | The molecular configuration of the CD20 of antigen contact or interactions with B cells and T cells
Nature Reviews
molecule. CD20 is a tetra-transmembrane | Nephrology
protein that
and are involved in chronic humoral autoimmunity and
essentially remains on the membrane of B cells without
dissociation or internalization upon antibody binding. treatment-refractory disease. The memory cell pool can
CD20 has small and large extracellular loops. The binding also be enriched by plasmablasts, which invade survival
sites of CD20 monoclonal antibodies, rituximab and niches in the bone marrow, lymph nodes, and inflamed
ofatumumab, are indicated. Rituximab binds an epitope tissue where they transform into long-lived memory
on the large loop only, whereas ofatumumab specifically plasma cells to sustain autoantibody production and
recognizes an epitope encompassing both the small and chronic inflammation. Speculatively, upon re‑exposure
large extracellular loops of the CD20 molecule. to antigens, memory cells could reactivate into plasma-
blasts and then convert to plasma cells to produce large
numbers of antibodies147.
In approximately half of patients for whom rituxi- In patients with membranous nephropathy, therefore,
mab therapy fails, the lack of success can be explained newly generated, short-lived plasmablasts attributable to
by the presence of severe and irreversible chronic renal B-cell hyperactivity are likely to produce the majority of
lesions at the time of treatment. However, in a small autoantibodies at disease onset and at the time of relapse,
but substantial subset of patients, rituximab seems to whereas memory plasma cells produced as a result of
fail because of ineffective or transient depletion of cir- autoreactive B-cell activation can sustain autoantibody
culating anti-PLA2R autoantibodies (and conceivably production despite therapy or disease relapses after initial
anti‑THSD7A or other unknown nephritogenic auto­ remission. Conceivably, both pathways might contrib-
antibodies)65. Some of these patients are prone to relapse. ute differently to autoantibody production in different
Three groups of patients can be identified according to patients and at different stages of the disease. Failure to
their clinical outcome. First, those who primarily fail achieve autoantibody depletion and disease remission
rituximab therapy. Second, those who initially respond can, therefore, be explained by two mechanisms that are
to treatment but then relapse. Third, those who, after not mutually exclusive — changes in the CD20 antigen
repeated exposures to rituximab, experience treatment that prevent B cell–rituximab binding and B-cell deple-
sensitization or an allergic reaction that can manifest as tion, and autoantibody production by long-lived memory
resistance to treatment or symptoms resembling serum plasma cells that do not express the CD20 antigen148.
sickness (for example, arthritis, arthralgias, myalgia,
and fever), which contraindicate further exposure to the Ineffective autoreactive B‑cell depletion. The cytotoxic
drug. With primary treatment failure and relapse, novel B-cell effect of rituximab seems to be mediated by its
treatments are needed to achieve antibody depletion by specific binding to the large loop of the CD20 antigen
mechanisms different from, or complementary to, those (FIG. 4) with secondary C1q capture, complement depo-
of rituximab. In patients with sensitization or allergic sition on B-cell membranes, and complement-mediated
reaction to treatment, humanized monoclonal anti­ cytolysis. The B-cell surface antigen, CD20, is a tetra-­
bodies that bind the CD20 antigen similarly to rituximab transmembrane protein involved in the phosphorylation
without inducing any immune response may be useful145. cascade of intracellular proteins and B-cell activation,
Autoantibodies against PLA2R, THSD7A, or other proliferation, and differentiation149. This protein pro-
unknown antigens, can be produced by two distinct anti- trudes from the surface of the B-cell membrane into
body-producing cell populations146: short-lived plasma the extracellular space by a small loop and a large loop,
cells and long-lived memory plasma cells. Short-lived which are binding sites for anti‑CD20 monoclonal anti-
plasma cells are usually generated in the spleen and lymph bodies85,150–152 (FIG. 4). As observed in treatment-­resistant
nodes from activated B cells. As observed after vaccina- B-cell lymphomas, some B cells become resistant to
tion or during episodes of immune-mediated disease, rituximab-induced complement-dependent cytotoxicity
these cells proliferate upon exposure to antigens and because of changes or internalization of the large loop of

574 | SEPTEMBER 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

the CD20 antigen, which prevents rituximab binding to Ofatumumab has been assessed as rituximab rescue
its specific epitope. To overcome this limitation of rituxi­ therapy in eleven patients with membranous nephro­
mab therapy, second and third generation anti‑CD20 pathy and without advanced renal insufficiency, and this
monoclonal antibodies, such as obinutuzumab and preliminary experi­ence is encouraging (P. Ruggenenti
ofatumumab, have been developed that bind different and G. Remuzzi; unpublished work; BOX 2). Blockade of
epitopes on the CD20 antigen. These agents have been factors or cells that stimu­late autoreactive B cells, such
proven effective for the treatment of B‑cell lymphomas153 as type I interferon, T-helper cells, or regulatory T cells,
(with some exceptions154) and other blood malignan- might also prevent the development of short-lived plas-
cies155 in patients who had previously failed rituximab mablasts and plasma cells160 (TABLE 2). A monoclonal anti-
therapy. Ofatumumab, unlike rituximab, specifically rec- body, belimumab, specifically targets the soluble form of
ognizes an epitope encompassing both the small and large B lymphocyte stimulator (BLyS, also known as BAFF),
extracellular loops of the CD20 molecule156 (FIG. 4), which a tumour necrosis factor super family ligand that has a
seems to be closely related to effective C1q capture and critical role in the differentiation and homeostasis of B
complement-­mediated cytotoxicity 155. The close binding lymphocytes. Via BLyS inhibition, belimumab might
proximity of ofatumumab to the cell membrane is likely to induce apoptosis and depletion of autoreactive B cells.
result in more efficient complement deposition on B‑cell The effects of belimumab on proteinuria and anti-PLA2R
membranes than achieved with rituximab, which might antibody production were evaluated in 14 patients with
explain the cytotoxic effects of this agent on rituximab-re- anti-PLA2R-positive membranous nephropathy and a
sistant B cells. Ofatumumab was approved by the FDA in urinary protein:creatinine ratio >400 mg/mmol from six
2009 and the European Medicines Agency in 2010 for the centres in the UK161. Treatment significantly and progres-
treatment of chronic lymphocytic leukaemia157 and could sively decreased anti-PLA2R antibody titre by week 12,
have additional indications for some non-Hodgkin lym- and reduced proteinuria together with normalization of
phomas and relapsing–remitting multiple sclerosis and serum albumin levels in patients with overt nephrotic
rheumatoid arthritis158. In 2014, ofatumumab was found syndrome. Of note, changes in anti-PLA2R antibody
to achieve complete disease remission in four children titre and in proteinuria seemed to parallel the changes
and a 19‑year old girl with focal segmental glomeru- observed after rituximab administration, but with an
losclerosis or minimal change disease ­associated with apparent delay of approximately 3–6 months (FIG. 5).
­rituximab-resistant nephrotic syndrome159. Conceivably, the faster effect of rituximab reflected
In patients with rituximab-resistant membranous immediate B-cell lysis, whereas the delayed effect of
nephropathy, rituximab fails to persistently deplete cir- belimumab might reflect progressive ‘exhaustion’ of
culating autoantibodies, but achieves the same deplet- antibody-producing B cells secondary to BLyS binding
ing effect on circulating B cells that is observed in and inhibition. Considering the different mechanisms
rituximab-­sensitive patients65. Therefore, changes in of B-cell inhibition, rituximab and belimumab given in
the CD20 antigen that prevent rituximab binding might combination could synergistically inhibit autoreactive
be restricted to autoreactive B-cell clones that continue B-cell clones162 and achieve faster and more efficient
to produce nephritogenic antibodies despite depletion inhibition of autoantibody production than rituximab
of the large majority of ‘innocent’ circulating B cells. or belimumab monotherapy.
New therapeutic options to inhibit the production of
autoantibodies could include interventions that specif-
Box 2 | Ofatumumab as rescue therapy in membranous nephropathy ically target the autoreactive B-cell clones involved in
the production of anti-PLA2R or anti‑THSD7A auto­
A 30‑year-old male patient who had failed a course of steroid and cyclophosphamide
according to KDIGO guidelines, achieved complete B‑cell depletion and proteinuria
antibodies, peptides that interact with the antibodies, or
remission following a single infusion of 375 mg/m2 of rituximab. After five relapses that antibody traps or decoys2,163.
invariably recovered after rituximab administration, the last rituximab infusion was Insights to aid the development of more targeted
ineffective. The outcome was complicated by fever and arthralgias, which were therapeutic approaches for the treatment of auto­
interpreted as signs of a serum-sickness-like syndrome. Circulating B cells were fully immune disease might be gleaned from a 2016 study
depleted after each rituximab infusion. He subsequently received one infusion of 300 mg of pemphigus vulgaris, an autoimmune disease of the
per m2 of body surface area of ofatumumab155 which, as previously observed with one skin mediated by IgG autoantibodies against desmo-
375 mg/1.73m2 infusion of rituximab119, achieved full B‑cell depletion within 24 h. glein 3 (REF. 164). Using an in vivo model of the disease,
Proteinuria progressively decreased up to complete remission of nephrotic syndrome researchers have engineered T cells to specifically rec-
over approximately 10 months. 18 months after administration of ofatumumab he was
ognize B cells that secrete anti-desmoglein 3 antibodies,
well, without proteinuria. A second patient who had failed a 6‑month steroid and
clorambucil course followed by 1-year ciclosporin therapy, received one infusion of
enabling their precise and efficient disposal164. Although
375 mg/m2 of rituximab. Despite depletion of circulating CD20 cells, proteinuria in its infancy, this pioneering approach could also ena-
continued to increase up to 15 g per 24 h and serum creatinine to 3.5 mg/dl ble antigen-specific immunosuppression in other B cell-­
(309.4 μmol/l). 300 mg of ofatumumab was subsequently infused. Treatment fully mediated autoimmune diseases for which autoantigens
depleted B cells from the circulation within 24 h, achieved complete and persistent have been identified, such as membranous nephropathy.
remission of nephrotic syndrome within 10 months, and even decreased serum
creatinine to pre-rituximab levels. 1‑year later the patient was well, without proteinuria Targeting memory plasma cells. Whether early-stage
and with near-normal serum creatinine levels. Thus, ofatumumab is a promising membranous nephropathy is predominantly mediated
alternative to rituximab and, cost effective, given that 300 mg of ofatumumab for a 70 kg by autoreactive B-cell clones sensitive to B‑cell targeted
patient costs €1,193 compared to €3,206 for 700 mg of rituximab120.
therapy, whereas more advanced disease is mediated by

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 575


©
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

a b Four 1.3 mg/m2 doses of bortezomib administered sub-


cutaneously at weekly intervals decreased autoantibody

Anti-PLA2R antibody titre (mean


percent change versus baseline)
percent change versus baseline)
ratio or 24 h proteinuria (mean

50 Rituximab 50 production and disease activity in 12 patients with cyclo-


Urinary protein to creatinine

Belimumab phosphamide and rituximab-resistant SLE170 and in one


0 0 patient with ANCA-associated vasculitis171. Bortezomib
has also been shown to prevent donor-specific antibody
production in kidney transplant recipients with anti-
–50 –50
body-mediated rejection172. A patient with membranous
nephropathy who had failed 6‑month treatment with
–100 –100 high-dose steroids achieved complete remission of the
0 3 6 9 12 18 24 0 3 6 9 12 18 24 nephrotic syndrome with bortezomib173, and another
Months Months patient with rituximab-resistant, post-transplantation
Figure 5 | Comparison of outcomes with belimumab or rituximab therapy. recurrent membranous nephropathy who was treated with
a | Percentage change in urinary protein to creatinine ratio Nature
(or 24 hReviews
proteinuria) versus
| Nephrology bortezomib achieved depletion of circulating CD34+ and
baseline. b | Percentage change in anti-M-type phospholipase A2 receptor (PLA2R) CD138+ cells and progressively reduced proteinuria over
antibody titre versus baseline. Data are from patients with membranous nephropathy 1 year 147. These findings indicate that plasma cells might
and nephrotic syndrome treated with belimumab (n = 14) or rituximab (n = 132)61.
be involved in the pathogenesis of primary and recurrent
Belimumab specifically targets the soluble form of B lymphocyte stimulator (BLyS,
also known as BAFF)161. Changes in proteinuria and anti-PLA2R antibody titre after
membranous nephropathy, particularly among patients
belimumab treatment seemed to parallel the changes observed after rituximab in whom anti‑CD20‑depletion therapy is not successful.
administration albeit with a delay in onset. Conceivably, the faster reduction in Unfortunately, bortezomib has serious adverse effects,
proteinuria and anti-PLA2R antibody titre after rituximab administration reflected the such as infections and neurotoxicity, which necessitate
immediate B-cell lysis achieved by treatment, whereas the slower effect of belimumab treatment interruption in most patients. The develop-
might reflect the progressive ‘exhaustion’ of antibody-producing B cells secondary to ment of second-generation proteasome inhibitors that
BLyS binding and inhibition. are as effective and safer than bortezomib, such as delan-
zomib and carfilzomib, will hopefully pave the way for
RCTs to evaluate the risk:benefit profile of this novel
autoreactive memory plasma cells resistant to anti-CD20 therapeutic option in patients with rituximab-resistant
monoclonal antibodies but sensitive to anti‑CD38 anti- membranous nephropathy.
bodies or proteosome inhibitors, is unknown. Moreover, Polyclonal antithymocyte globulins (ATGs) are often
whether transition from a process predominantly medi- used in induction immunosuppressive protocols to pre-
ated by B cells to one that is predominantly mediated by vent rejection of solid allografts or autologous haemato-
plasma cells is, to some extent, associated with epitope poietic stem cells. ATGs bind to antigens such as CD38,
spreading and the development of anti‑CTLD1 and CD138, and CD52, which are expressed on plasma
anti‑CTLD7 autoreactivity is also unknown, but is an cells, including autoreactive memory plasma cells174. In
intriguing proposal that might explain why patients with patients with severe autoimmune diseases refractory to
epitope spreading do not benefit from B cell-­targeted conventional immunosuppression, ATGs could achieve
therapy. Memory plasma cells survive rituximab ther- complete and sustained remission. The process would
apy because they do not express the CD20 antigen and involve an ‘immunoablation’ followed by the reconstitu-
continue to produce autoantibodies despite effective tion of an ‘adapted’ immune system devoid of autoreactive
B‑cell depletion from the circulation148,165. Plasma cells, long-lived memory plasma cells and which is, therefore,
however, strongly express CD38, a multifunctional cell self-tolerant175. This immunoablation might explain why
surface protein that is expressed at low levels in other treatment-resistant membranous nephropathy does not
blood cells and solid tissues166. CD38‑positive plasma recur in many patients who have undergone kidney trans-
cells can enrich chronically inflamed tissues and secrete plantation, and why anti‑CD20 targeted therapy achieves
autoantibodies165. Autoreactive plasma cells could there- remission of membranous nephropathy in patients with
fore be a target for anti‑CD38 monoclonal antibodies, kidney grafts more effectively than in native kidneys.
such as daratumumab and isatuximab. Like anti‑CD20 Unfortunately, the potentially devastating effects of even
antibodies that were initially introduced for the treatment a transient ablation of the immune system do not seem
of B‑cell lymphomas, these agents have been developed to justify this therapeutic approach for the treatment of
to kill malignant plasma cells166 and could be tested in membranous nephropathy. More-selective approaches
patients with plasma cell-mediated autoimmune diseases, under investigation explore the possibility of inhibiting
including some forms of membranous nephropathy. adhesion molecules, which contribute to plasma cell
Other molecules, such as the proteasome inhibitor homing and survival in the bone marrow niche and
bortezomib, have been reported to effectively deplete recruitment of inflammatory cells into the bone marrow
plasma cells, particularly activated plasma cells that, or inflamed tissue. Ideally, novel drugs should specifically
during forced production of circulating autoanti­bodies, disrupt autoreactive plasma cells without affecting plasma
are exposed to unfolded protein accumulation and cells that secrete protective antibodies146.
consequent apoptosis167. Bortezomib depletes auto­
reactive plasma cells and reduces ANCA and anti-double Targeting complement. Complement is activated by anti-
stranded DNA antibody titres in experimental models body interaction with target antigens on the podocyte
of ANCA-associated nephritis168 or lupus nephritis169. and at least part of the glomerular injury initiated by this

576 | SEPTEMBER 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

interaction is complement-mediated (FIG. 2). Complement evaluation of serum autoantibody titre and proteinuria,
could, therefore, be another target for the treatment of together with serum albumin levels in patients with overt
membranous nephropathy. One, unfortunately unpub- nephrotic syndrome could guide diagnosis of membra-
lished, prospective, randomized, clinical trial did not nous nephropathy and individually tailored treatment
demonstrate any antiproteinuric effect of 4 month ecu- protocols. Conventional, nonspecific, and toxic immuno-
lizumab therapy (8 mg/kg every 2 or 4 weeks) compared suppressive regimens will become treatments of the past6,
to placebo in 117 patients with membranous nephro­ to be replaced by more disease-specific and safer medica-
pathy. The results of the study were, however, incon- tions, such as B cell-targeting monoclonal antibodies and
clusive because underdosing of eculizumab resulted in autoreactive plasma cell inhibitors, paving the way for a
ineffective complement inhibition176. Novel complement novel therapeutic paradigm based on the principles of
inhibitors including oral agents, recombinant proteins, precision medicine and personalized therapy 9. Complex
small molecules, new monoclonal antibodies, small predictive models to identify which patients to treat and
interfering RNA agents, and approaches that upregulate when179 will no longer be needed, and even the most frail
natural complement inhibitors will become available for patients with severe disease might benefit from new, safe
clinical use in the near future. These agents are likely therapeutic options. Treatment could be anticipated at
to be evaluated in new trials of complement-targeting earlier and milder stages of membranous nephropathy,
interventions, particularly in patients with membranous even before the onset of nephrotic syndrome, to pre-
nephropathy and nephrotic syndrome in whom CD20 vent chronic and potentially irreversible histological
targeted therapy fails163,177,178. changes78. Patients who were previously left untreated,
so as not to expose them to the toxicity of cyclophospha-
Conclusions mide, might benefit from B cell-targeted therapy, which
Over the past decade, major advances in the understand- can prevent memory cell clone expansion associated with
ing of the mechanisms of membranous nephropathy have uncontrolled disease progression towards disease that is
led to novel perspectives on treatment. The integrated refractory to currently available therapy 146.

1. McQuarrie, E. P., Mackinnon, B., Stewart, G. A. & losartan in patients with idiopathic membranous 29. Perna, A. et al. Immunosuppressive treatment for
Geddes, C. C. Membranous nephropathy remains nephropathy and nephrotic syndrome. Scand. J. Urol. idiopathic membranous nephropathy: a systematic
the commonest primary cause of nephrotic syndrome Nephrol. 44, 251–256 (2010). review. Am. J. Kidney Dis. 44, 385–401 (2004).
in a northern European Caucasian population. 15. du Buf-Vereijken, P. W., Branten, A. J. & Wetzels, J. F. 30. Davison, A. M., Cameron, J. S., Kerr, D. N., Ogg, C. S.
Nephrol. Dial. Transplant. 25, 1009–1010 (2010). Idiopathic membranous nephropathy: outline and & Wilkinson, R. W. The natural history of renal
2. Ronco, P. & Debiec, H. Pathophysiological advances in rationale of a treatment strategy. Am. J. Kidney Dis. function in untreated idiopathic membranous
membranous nephropathy: time for a shift in patient’s 46, 1012–1029 (2005). glomerulonephritis in adults. Clin. Nephrol. 22, 61–67
care. Lancet 385, 1983–1992 (2015). 16. Troyanov, S., Wall, C. A., Miller, J. A., Scholey, J. W. & (1984).
3. Cattran, D. C., Kim, E. D., Reich, H., Hladunewich, M. Cattran, D. C. Idiopathic membranous nephropathy: 31. Chen, Y. et al. Immunosuppression for membranous
& Kim, S. J. Membranous nephropathy: quantifying definition and relevance of a partial remission. nephropathy: a systematic review and meta-analysis of
remission duration on outcome. J. Am. Soc. Nephrol. Kidney Int. 66, 1199–1205 (2004). 36 clinical trials. Clin. J. Am. Soc. Nephrol. 8, 787–796
28, 995–1003 (2017). 17. Praga, M. et al. Nephrotic proteinuria without (2013).
4. Ruggenenti, P., Perna, A. & Remuzzi, G. Retarding hypoalbuminemia: clinical characteristics and 32. Ponticelli, C. et al. A randomized study comparing
progression of chronic renal disease: the neglected response to angiotensin-converting enzyme inhibition. methylprednisolone plus chlorambucil versus
issue of residual proteinuria. Kidney Int. 63, Am. J. Kidney Dis. 17, 330–338 (1991). methylprednisolone plus cyclophosphamide in
2254–2261 (2003). 18. Praga, M. et al. Long-term beneficial effects of idiopathic membranous nephropathy. J. Am. Soc.
5. Schieppati, A., Ruggenenti, P., Perna, A. & Remuzzi, G. angiotensin-converting enzyme inhibition in patients Nephrol. 9, 444–450 (1998).
Nonimmunosuppressive therapy of membranous with nephrotic proteinuria. Am. J. Kidney Dis. 20, 33. Howman, A. et al. Immunosuppression for progressive
nephropathy. Semin. Nephrol. 23, 333–339 (2003). 240–248 (1992). membranous nephropathy: a UK randomised
6. Glassock, R. J. The treatment of idiopathic 19. Cravedi, P. et al. Spironolactone plus full-dose ACE controlled trial. Lancet 381, 744–751 (2013).
membranous nephropathy: a dilemma or a inhibition in patients with idiopathic membranous 34. Ponticelli, C. et al. A 10‑year follow‑up of a randomized
conundrum? Am. J. Kidney Dis. 44, 562–566 (2004). nephropathy and nephrotic syndrome: does it really study with methylprednisolone and chlorambucil in
7. Schieppati, A. et al. Prognosis of untreated patients work? Pharmaceuticals (Basel) 3, 1–9 (2010). membranous nephropathy. Kidney Int. 48, 1600–1604
with idiopathic membranous nephropathy. 20. Pisoni, R. et al. Effect of high dose ramipril with or (1995).
N. Engl. J. Med. 329, 85–89 (1993). without indomethacin on glomerular selectivity. 35. Ponticelli, C. et al. A randomized trial of
8. Garattini, S., Bertani, T. & Remuzzi, G. Steroids and Kidney Int. 62, 1010–1019 (2002). methylprednisolone and chlorambucil in idiopathic
cytotoxic drugs in the treatment of membranous 21. Campbell, R. et al. Effects of combined ACE inhibitor membranous nephropathy. N. Engl. J. Med. 320,
glomerulopathy. Nephron 50, 263–264 (1988). and angiotensin II antagonist treatment in human 8–13 (1989).
9. Mirnezami, R., Nicholson, J. & Darzi, A. Preparing for chronic nephropathies. Kidney Int. 63, 1094–1103 36. Essential Medicines and Health Products Information
precision medicine. N. Engl. J. Med. 366, 489–491 (2003). Portal. Guideline for good clinical practice. ICH
(2012). 22. Fervenza, F. C. et al. Rituximab therapy in idiopathic harmonised tripartite guideline. E6(R1). Current step 4
10. KDIGO. KDIGO clinical practice guidelines for membranous nephropathy: a 2‑year study. Clin. J. Am. version, dated 10 June 1996. (including the post step 4
glomerulonephritis. Chapter 7: idiopathic Soc. Nephrol. 5, 2188–2198 (2010). corrections). The International Conference on
membranous nephropathy. Kidney Int. Suppl. 2, 23. Fervenza, F. C. et al. Rituximab treatment of idiopathic Harmonization of Technical Requirements for
186–197 (2012). membranous nephropathy. Kidney Int. 73, 117–125 Registration of Pharmaceuticals for Human Use (ICH).
11. Remuzzi, G., Benigni, A. & Remuzzi, A. Mechanisms of (2008). http://apps.who.int/medicinedocs/en/m/abstract/
progression and regression of renal lesions of chronic 24. Ruggenenti, P. et al. Diverse effects of increasing Js22154en/ (1996).
nephropathies and diabetes. J. Clin. Invest. 116, lisinopril doses on lipid abnormalities in chronic 37. Saxena, P. & Saxena, R. Clinical trials: changing
288–296 (2006). nephropathies. Circulation 107, 586–592 (2003). regulations in India. Indian J. Community Med. 39,
12. Ruggenenti, P. et al. ACE inhibition improves 25. Dahan, K. et al. Rituximab for severe membranous 197–202 (2014).
glomerular size selectivity in patients with idiopathic nephropathy: a 6‑month trial with extended follow‑up. 38. Jha, V. et al. A randomized, controlled trial of steroids
membranous nephropathy and persistent nephrotic J. Am. Soc. Nephrol. 28, 348–358 (2017). and cyclophosphamide in adults with nephrotic
syndrome. Am. J. Kidney Dis. 35, 381–391 (2000). 26. Ruggenenti, P., Schieppati, A. & Remuzzi, G. syndrome caused by idiopathic membranous
13. Gansevoort, R. T., Heeg, J. E., Vriesendorp, R., de Progression, remission, regression of chronic renal nephropathy. J. Am. Soc. Nephrol. 18, 1899–1904
Zeeuw, D. & de Jong, P. E. Antiproteinuric drugs in diseases. Lancet 357, 1601–1608 (2001). (2007).
patients with idiopathic membranous glomerulopathy. 27. Ruggenenti, P., Brenner, B. M. & Remuzzi, G. 39. Faurschou, M. et al. Malignancies in Wegener’s
Nephrol. Dial. Transplant. 7 (Suppl. 1), 91–96 Remission achieved in chronic nephropathy by a granulomatosis: incidence and relation to
(1992). multidrug approach targeted at urinary protein cyclophosphamide therapy in a cohort of 293 patients.
14. Kosmadakis, G., Filiopoulos, V., Georgoulias, C., excretion. Nephron 88, 254–259 (2001). J. Rheumatol. 35, 100–105 (2008).
Tentolouris, N. & Michail, S. Comparison of the 28. Ruggenenti, P. et al. Role of remission clinics in the 40. Ruggenenti, P. & Remuzzi, G. Idiopathic membranous
influence of angiotensin-converting enzyme inhibitor longitudinal treatment of CKD. J. Am. Soc. Nephrol. nephropathy: back to the future? Lancet 381, 706–708
lisinopril and angiotensin II receptor antagonist 19, 1213–1224 (2008). (2013).

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 577


©
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

41. van den Brand, J. et al. Safety of rituximab compared membranous nephropathy. J. Am. Soc. Nephrol. 26, a prospective, matched-cohort study. Am. J. Nephrol.
with steroids and cyclophosphamide for idiopathic 2545–2558 (2015). 33, 461–468 (2011).
membranous nephropathy. J. Am. Soc. Nephrol. 66. Hofstra, J. M. et al. Antiphospholipase A2 receptor 89. Segarra, A. et al. Successful treatment of membranous
http://dx.doi.org/10.1681/ASN.2016091022 (2017). antibody titer and subclass in idiopathic membranous glomerulonephritis with rituximab in calcineurin
42. Remuzzi, G. et al. Rituximab for idiopathic membranous nephropathy. J. Am. Soc. Nephrol. 23, 1735–1743 inhibitor-dependent patients. Clin. J. Am. Soc. Nephrol.
nephropathy. Lancet 360, 923–924 (2002). (2012). 4, 1083–1088 (2009).
43. Beck, L. H. Jr et al. M‑Type phospholipase A2 receptor 67. Timmermans, S. A., Abdul Hamid, M. A., Cohen 90. Bariety, J., Samarcq, P., Lagrue, G., Fritel, D. &
as target antigen in idiopathic membranous Tervaert, J. W., Damoiseaux, J. G. & van Paassen, P. Milliez, P. Recovery of normal ultrastructure in 2 cases
nephropathy. N. Engl. J. Med. 361, 11–21 (2009). Anti‑PLA2R antibodies as a prognostic factor in of primary glomerulonephritis with diffuse extra-
44. Appel, G. B. Rituximab in membranous nephropathy: PLA2R‑related membranous nephropathy. membranous deposits [French]. Presse Med. 76,
is it a first-line treatment? J. Am. Soc. Nephrol. 23, Am. J. Nephrol. 42, 70–77 (2015). 2179–2182 (1968).
1280–1282 (2012). 68. Kanigicherla, D. et al. Anti‑PLA2R antibodies measured 91. Forland, M. & Spargo, B. H. Clinicopathological
45. van den Brand, J. A., van Dijk, P. R., Hofstra, J. M. by ELISA predict long-term outcome in a prevalent correlations in idiopathic nephrotic syndrome with
& Wetzels, J. F. Long-term outcomes in idiopathic population of patients with idiopathic membranous membranous nephropathy. Nephron 6, 498–525
membranous nephropathy using a restrictive treatment nephropathy. Kidney Int. 83, 940–948 (2013). (1969).
strategy. J. Am. Soc. Nephrol. 25, 150–158 (2014). 69. Radice, A. et al. Clinical usefulness of autoantibodies to 92. Osawa, G., Kinoshita, Y., Morita, T., Soda, T. & Wada, J.
46. van den Brand, J. A., van Dijk, P. R., Hofstra, J. M. M‑type phospholipase A2 receptor (PLA2R) for Resolution of diffuse membranous changes with
& Wetzels, J. F. Cancer risk after cyclophosphamide monitoring disease activity in idiopathic membranous remission of nephrotic syndrome. Tohoku J. Exp. Med.
treatment in idiopathic membranous nephropathy. nephropathy (IMN). Autoimmun. Rev. 15, 146–154 120, 177–190 (1976).
Clin. J. Am. Soc. Nephrol. 9, 1066–1073 (2014). (2016). 93. Ambalavanan, S., Fauvel, J. P., Sibley, R. K. &
47. van de Logt, A. E., Hofstra, J. M. & Wetzels, J. F. 70. Hoxha, E. et al. An indirect immunofluorescence Myers, B. D. Mechanism of the antiproteinuric effect of
Pharmacological treatment of primary membranous method facilitates detection of thrombospondin type 1 cyclosporine in membranous nephropathy. J. Am. Soc.
nephropathy in 2016. Expert Rev. Clin. Pharmacol. 9, domain-containing 7A‑specific antibodies in Nephrol. 7, 290–298 (1996).
1463–1478 (2016). membranous nephropathy. J. Am. Soc. Nephrol. 28, 94. Marasa, M., Cravedi, P., Ruggiero, B. & Ruggenenti, P.
48. Hofstra, J. M., Fervenza, F. C. & Wetzels, J. F. 520–531 (2017). Refractory focal segmental glomerulosclerosis in the
Treatment of idiopathic membranous nephropathy. 71. Ancian, P., Lambeau, G., Mattei, M. G. & adult: complete and sustained remissions of two
Nat. Rev. Nephrol. 9, 443–458 (2013). Lazdunski, M. The human 180‑kDa receptor for episodes of nephrotic syndrome after a single dose of
49. Jones, D. B. Nephrotic glomerulonephritis. secretory phospholipases A2. Molecular cloning, rituximab. BMJ Case Rep. http://dx.doi.org/10.1136/
Am. J. Pathol. 33, 313–329 (1957). identification of a secreted soluble form, expression, bcr-2014-205507 (2014).
50. Heymann, W., Hackel, D. B., Harwood, S., Wilson, S. G. and chromosomal localization. J. Biol. Chem. 270, 95. Meijer, J. M. et al. Effectiveness of rituximab treatment
& Hunter, J. L. Production of nephrotic syndrome in 8963–8970 (1995). in primary Sjogren’s syndrome: a randomized, double-
rats by Freund’s adjuvants and rat kidney suspensions. 72. Fresquet, M. et al. Identification of a major epitope blind, placebo-controlled trial. Arthritis Rheum. 62,
Proc. Soc. Exp. Biol. Med. 100, 660–664 (1959). recognized by PLA2R autoantibodies in primary 960–968 (2010).
51. Kerjaschki, D. & Farquhar, M. G. The pathogenic membranous nephropathy. J. Am. Soc. Nephrol. 26, 96. Dass, S. et al. Reduction of fatigue in Sjogren syndrome
antigen of Heymann nephritis is a membrane 302–313 (2015). with rituximab: results of a randomised, double-blind,
glycoprotein of the renal proximal tubule brush border. 73. Seitz-Polski, B. et al. Epitope spreading of placebo-controlled pilot study. Ann. Rheum. Dis. 67,
Proc. Natl Acad. Sci. USA 79, 5557–5561 (1982). autoantibody response to PLA2R associates with poor 1541–1544 (2008).
52. Debiec, H. et al. Antenatal membranous prognosis in membranous nephropathy. J. Am. Soc. 97. Merrill, J. T. et al. Efficacy and safety of rituximab in
glomerulonephritis due to anti-neutral endopeptidase Nephrol. 27, 1517–1533 (2016). moderately-to‑severely active systemic lupus
antibodies. N. Engl. J. Med. 346, 2053–2060 (2002). 74. van der Woude, D. et al. Gene-environment interaction erythematosus: the randomized, double-blind, phase II/
53. Hofstra, J. M. et al. Anti-phospholipase A2 receptor influences the reactivity of autoantibodies to III systemic lupus erythematosus evaluation of
antibodies correlate with clinical status in idiopathic citrullinated antigens in rheumatoid arthritis. rituximab trial. Arthritis Rheum. 62, 222–233 (2010).
membranous nephropathy. Clin. J. Am. Soc. Nephrol. Nat. Genet. 42, 814–816 (2010). 98. Stone, J. H. et al. Rituximab versus cyclophosphamide
6, 1286–1291 (2011). 75. Shah, P., Tramontano, A. & Makker, S. P. Intramolecular for ANCA-associated vasculitis. N. Engl. J. Med. 363,
54. Qin, W. et al. Anti-phospholipase A2 receptor antibody epitope spreading in Heymann nephritis. J. Am. Soc. 221–232 (2010).
in membranous nephropathy. J. Am. Soc. Nephrol. 22, Nephrol. 18, 3060–3066 (2007). 99. Jones, R. B. et al. Rituximab versus cyclophosphamide
1137–1143 (2011). 76. Ruggenenti, P. et al. Effects of rituximab on in ANCA-associated renal vasculitis. N. Engl. J. Med.
55. Stanescu, H. C. et al. Risk HLA‑DQA1 and PLA2R1 morphofunctional abnormalities of membranous 363, 211–220 (2010).
alleles in idiopathic membranous nephropathy. glomerulopathy. Clin. J. Am. Soc. Nephrol. 3, 100. Carson, K. R. et al. Progressive multifocal
N. Engl. J. Med. 364, 616–626 (2011). 1652–1659 (2008). leukoencephalopathy after rituximab therapy in HIV-
56. Tomas, N. M. et al. Thrombospondin type‑1 domain- 77. Cravedi, P., Remuzzi, G. & Ruggenenti, P. Rituximab in negative patients: a report of 57 cases from the
containing 7A in idiopathic membranous nephropathy. primary membranous nephropathy: first-line therapy, Research on Adverse Drug Events and Reports project.
N. Engl. J. Med. 371, 2277–2287 (2014). why not? Nephron Clin. Pract. 128, 261–269 (2014). Blood 113, 4834–4840 (2009).
57. Larsen, C. P., Cossey, L. N. & Beck, L. H. THSD7A 78. De Vriese, A. S., Glassock, R. J., Nath, K. A., Sethi, S. 101. Molloy, E. S. & Calabrese, L. H. Progressive multifocal
staining of membranous glomerulopathy in clinical & Fervenza, F. C. A proposal for a serology-based leukoencephalopathy: a national estimate of frequency
practice reveals cases with dual autoantibody approach to membranous nephropathy. J. Am. Soc. in systemic lupus erythematosus and other rheumatic
positivity. Mod. Pathol. 29, 421–426 (2016). Nephrol. 28, 421–430 (2017). diseases. Arthritis Rheum. 60, 3761–3765 (2009).
58. Lv, J. et al. Interaction between PLA2R1 and 79. Debiec, H. & Ronco, P. Immunopathogenesis of 102. Pugnet, G. et al. Progressive multifocal encephalopathy
HLA‑DQA1 variants associates with anti‑PLA2R membranous nephropathy: an update. Semin. after cyclophosphamide in granulomatosis with
antibodies and membranous nephropathy. J. Am. Soc. Immunopathol. 36, 381–397 (2014). polyangiitis (Wegener) patients: case report and review
Nephrol. 24, 1323–1329 (2013). 80. Cupps, T. R., Edgar, L. C. & Fauci, A. S. Suppression of of literature. Clin. Exp. Rheumatol. 31, S62–64 (2013).
59. Beck, L. H. Jr et al. Rituximab-induced depletion of human B lymphocyte function by cyclophosphamide. 103. Sellier-Leclerc, A. L., Belli, E., Guerin, V., Dorfmuller, P.
anti‑PLA2R autoantibodies predicts response in J. Immunol. 128, 2453–2457 (1982). & Deschenes, G. Fulminant viral myocarditis after
membranous nephropathy. J. Am. Soc. Nephrol. 22, 81. Pescovitz, M. D. Rituximab, an anti‑cd20 monoclonal rituximab therapy in pediatric nephrotic syndrome.
1543–1550 (2011). antibody: history and mechanism of action. Pediatr. Nephrol. 28, 1875–1879 (2013).
60. Oh, Y. J., Yang, S. H., Kim, D. K., Kang, S. W. & Am. J. Transplant. 6, 859–866 (2006). 104. Delbue, S. et al. Investigation of polyomaviruses
Kim, Y. S. Autoantibodies against phospholipase A2 82. Ruggenenti, P. et al. Rituximab in idiopathic replication in pediatric patients with nephropathy
receptor in Korean patients with membranous membranous nephropathy: a one-year prospective receiving rituximab. J. Med. Virol. 84, 1464–1470
nephropathy. PLoS ONE 8, e62151 (2013). study. J. Am. Soc. Nephrol. 14, 1851–1857 (2003). (2012).
61. Bech, A. P., Hofstra, J. M., Brenchley, P. E. & 83. Bomback, A. S. et al. Rituximab therapy for 105. Iijima, K. et al. Rituximab for childhood-onset,
Wetzels, J. F. Association of anti-PLA2R antibodies membranous nephropathy: a systematic review. complicated, frequently relapsing nephrotic syndrome
with outcomes after immunosuppressive therapy in Clin. J. Am. Soc. Nephrol. 4, 734–744 (2009). or steroid-dependent nephrotic syndrome: a
idiopathic membranous nephropathy. Clin. J. Am. Soc. 84. Piro, L. D. et al. Extended rituximab (anti‑CD20 multicentre, double-blind, randomised, placebo-
Nephrol. 9, 1386–1392 (2014). monoclonal antibody) therapy for relapsed or controlled trial. Lancet 384, 1273–1281 (2014).
62. Hoxha, E. et al. Phospholipase A2 receptor refractory low-grade or follicular non-Hodgkin’s 106. Ruggenenti, P. et al. Rituximab in steroid-dependent or
autoantibodies and clinical outcome in patients with lymphoma. Ann. Oncol. 10, 655–661 (1999). frequently relapsing idiopathic nephrotic syndrome.
primary membranous nephropathy. J. Am. Soc. 85. McLaughlin, P. et al. Rituximab chimeric anti‑CD20 J. Am. Soc. Nephrol. 25, 850–863 (2014).
Nephrol. 25, 1357–1366 (2014). monoclonal antibody therapy for relapsed indolent 107. Gea-Banacloche, J. C. Rituximab-associated infections.
63. Hoxha, E., Harendza, S., Pinnschmidt, H., Panzer, U. lymphoma: half of patients respond to a four-dose Semin. Hematol. 47, 187–198 (2010).
& Stahl, R. A. M‑Type phospholipase A2 receptor treatment program. J. Clin. Oncol. 16, 2825–2833 108. Lanini, S. et al. Risk of infection in patients with
autoantibodies and renal function in patients with (1998). lymphoma receiving rituximab: systematic review and
primary membranous nephropathy. Clin. J. Am. Soc. 86. Ruggenenti, P. et al. Rituximab in idiopathic meta-analysis. BMC Med. 9, 36 (2011).
Nephrol. 9, 1883–1890 (2014). membranous nephropathy. J. Am. Soc. Nephrol. 23, 109. Tan, C. S. & Koralnik, I. J. Progressive multifocal
64. Hoxha, E., Harendza, S., Pinnschmidt, H., Panzer, U. 1416–1425 (2012). leukoencephalopathy and other disorders caused by JC
& Stahl, R. A. PLA2R antibody levels and clinical 87. Makker, S. P. & Kanalas, J. J. Course of transplanted virus: clinical features and pathogenesis. Lancet Neurol.
outcome in patients with membranous nephropathy Heymann nephritis kidney in normal host. Implications 9, 425–437 (2010).
and non-nephrotic range proteinuria under treatment for mechanism of proteinuria in membranous 110. van Vollenhoven, R. F., Fleischmann, R. M., Furst, D. E.,
with inhibitors of the renin-angiotensin system. glomerulonephropathy. J. Immunol. 142, 3406–3410 Lacey, S. & Lehane, P. B. Longterm safety of rituximab:
PLoS ONE 9, e110681 (2014). (1989). final report of the rheumatoid arthritis global clinical
65. Ruggenenti, P. et al. Anti-phospholipase A2 receptor 88. Cravedi, P. et al. Efficacy and safety of rituximab trial program over 11 years. J. Rheumatol. 42,
antibody titer predicts post-rituximab outcome of second-line therapy for membranous nephropathy: 1761–1766 (2015).

578 | SEPTEMBER 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

111. Jemal, A. et al. Cancer statistics, 2006. CA Cancer membranous nephropathy after kidney transplantation: lupus nephritis. J. Pharmacol. Pharmacother. 6, 71–76
J. Clin. 56, 106–130 (2006). a surveillance biopsy study. Am. J. Transplant. 8, (2015).
112. Feehally, J. Targeted therapies: is there a role for 1318–1322 (2008). 161. Willcocks, L. et al. Effect of belimumab on proteinuria
rituximab in nephrotic syndrome? Nat. Rev. Nephrol. 136. El‑Zoghby, Z. M. et al. Recurrent idiopathic and anti‑PLA2R autoantibody in idiopathic
10, 245–247 (2014). membranous nephropathy: early diagnosis by protocol membranous nephropathy — 6 months data.
113. Tavazzi, E., Ferrante, P. & Khalili, K. Progressive biopsies and treatment with anti‑CD20 monoclonal Nephrol. Dial. Transplant. 30, iii32–iii33 (2015).
multifocal leukoencephalopathy: an unexpected antibodies. Am. J. Transplant. 9, 2800–2807 (2009). 162. Gong, Q. et al. Importance of cellular microenvironment
complication of modern therapeutic monoclonal 137. Marcen, R. et al. Membranous nephropathy: and circulatory dynamics in B cell immunotherapy.
antibody therapies. Clin. Microbiol. Infect. 17, recurrence after kidney transplantation. Nephrol. Dial. J. Immunol. 174, 817–826 (2005).
1776–1780 (2011). Transplant. 11, 1129–1133 (1996). 163. Debiec, H. & Ronco, P. Immune response against
114. Clifford, D. B. et al. Rituximab-associated progressive 138. Poduval, R. D., Josephson, M. A. & Javaid, B. Treatment autoantigen PLA2R is not gambling: implications for
multifocal leukoencephalopathy in rheumatoid of de novo and recurrent membranous nephropathy in pathophysiology, prognosis, and therapy. J. Am. Soc.
arthritis. Arch. Neurol. 68, 1156–1164 (2011). renal transplant patients. Semin. Nephrol. 23, 392–399 Nephrol. 27, 1275–1277 (2016).
115. Kaufman, G. P. et al. Progressive multifocal (2003). 164. Ellebrecht, C. T. et al. Reengineering chimeric antigen
leukoencephalopathy: a rare infectious complication 139. Rodriguez, E. F. et al. The pathology and clinical receptor T cells for targeted therapy of autoimmune
following allogeneic hematopoietic cell features of early recurrent membranous disease. Science 353, 179–184 (2016).
transplantation (HCT). Eur. J. Haematol. 92, 83–87 glomerulonephritis. Am. J. Transplant. 12, 1029–1038 165. Mei, H. E. et al. A unique population of IgG-expressing
(2014). (2012). plasma cells lacking CD19 is enriched in human bone
116. Ruggenenti, P., Cravedi, P. & Remuzzi, G. Rituximab 140. Grupper, A. et al. Recurrent membranous nephropathy marrow. Blood 125, 1739–1748 (2015).
for membranous nephropathy and immune disease: after kidney transplantation: treatment and long-term 166. van de Donk, N. W. et al. Monoclonal antibodies
less might be enough. Nat. Clin. Pract. Nephrol. 5, implications. Transplantation 100, 2710–2716 targeting CD38 in hematological malignancies and
76–77 (2009). (2016). beyond. Immunol. Rev. 270, 95–112 (2016).
117. Gabardi, S., Martin, S. T., Roberts, K. L. & Grafals, M. 141. Kattah, A. et al. Anti-phospholipase A2 receptor 167. Meister, S. et al. Extensive immunoglobulin production
Induction immunosuppressive therapies in renal antibodies in recurrent membranous nephropathy. sensitizes myeloma cells for proteasome inhibition.
transplantation. Am. J. Health Syst. Pharm. 68, Am. J. Transplant. 15, 1349–1359 (2015). Cancer Res. 67, 1783–1792 (2007).
211–218 (2011). 142. Quintana, L. F. et al. Antiphospholipase A2 receptor 168. Bontscho, J. et al. Myeloperoxidase-specific plasma cell
118. Pepper, R. et al. Rituximab is an effective treatment antibody levels predict the risk of posttransplantation depletion by bortezomib protects from anti-neutrophil
for lupus nephritis and allows a reduction in recurrence of membranous nephropathy. cytoplasmic autoantibodies-induced
maintenance steroids. Nephrol. Dial. Transplant. 24, Transplantation 99, 1709–1714 (2015). glomerulonephritis. J. Am. Soc. Nephrol. 22, 336–348
3717–3723 (2009). 143. El‑Zoghby, Z. M. et al. Identifying specific causes of (2011).
119. Cravedi, P., Ruggenenti, P., Sghirlanzoni, M. C. & kidney allograft loss. Am. J. Transplant. 9, 527–535 169. Neubert, K. et al. The proteasome inhibitor bortezomib
Remuzzi, G. Titrating rituximab to circulating B cells to (2009). depletes plasma cells and protects mice with lupus-like
optimize lymphocytolytic therapy in idiopathic 144. Sellares, J. et al. Understanding the causes of kidney disease from nephritis. Nat. Med. 14, 748–755
membranous nephropathy. Clin. J. Am. Soc. Nephrol. transplant failure: the dominant role of antibody- (2008).
2, 932–937 (2007). mediated rejection and nonadherence. 170. Alexander, T. et al. The proteasome inhibitior
120. Agenzia Italiana del Farmaco [Italian]. Gazzetta Am. J. Transplant. 12, 388–399 (2012). bortezomib depletes plasma cells and ameliorates
Ufficiale http://95.110.157.84/gazzettaufficiale.biz/ 145. Vivarelli, M. et al. Ofatumumab in two pediatric clinical manifestations of refractory systemic lupus
atti/2012/20120013/12A00419.htm (2011). nephrotic syndrome patients allergic to rituximab. erythematosus. Ann. Rheum. Dis. 74, 1474–1478
121. Medindia. Drug “cyclophosphamide” price list. Pediatr. Nephrol. 32, 181–184 (2017). (2015).
Medindia http://www.medindia.net/drug-price/ 146. Hiepe, F. & Radbruch, A. Plasma cells as an innovative 171. Novikov, P., Moiseev, S., Bulanov, N. & Shchegoleva, E.
cyclophosphamide.htm (updated 2 Feb 2017). target in autoimmune disease with renal Bortezomib in refractory ANCA-associated vasculitis:
122. ASST‑PG23. Dialysis costs http://www.asst-pg23.it/ manifestations. Nat. Rev. Nephrol. 12, 232–240 a new option? Ann. Rheum. Dis. 75, e9 (2016).
component/trasparenza/12 (2017). (2016). 172. Ejaz, N. S. et al. Review of bortezomib treatment of
123. Goodrx. Tacrolimus https://www.goodrx.com/ 147. Barbari, A. et al. Bortezomib as a novel approach to antibody-mediated rejection in renal transplantation.
tacrolimus (2017). early recurrent membranous glomerulonephritis after Antioxid. Redox Signal. 21, 2401–2418 (2014).
124. Cattran, D. C. et al. Cyclosporine in patients with kidney transplant refractory to combined conventional 173. Hartono, C., Chung, M., Kuo, S. F., Seshan, S. V. &
steroid-resistant membranous nephropathy: rituximab therapy. Exp. Clin. Transplant. 15, 350–354 Muthukumar, T. Bortezomib therapy for nephrotic
a randomized trial. Kidney Int. 59, 1484–1490 (2017). syndrome due to idiopathic membranous nephropathy.
(2001). 148. Hiepe, F. et al. Long-lived autoreactive plasma cells J. Nephrol. 27, 103–106 (2014).
125. Praga, M., Barrio, V., Juarez, G. F. & Luno, J. drive persistent autoimmune inflammation. Nat. Rev. 174. Popow, I. et al. A comprehensive and quantitative
Tacrolimus monotherapy in membranous Rheumatol. 7, 170–178 (2011). analysis of the major specificities in rabbit
nephropathy: a randomized controlled trial. 149. Uchiyama, S. et al. Development of novel humanized antithymocyte globulin preparations. Am. J. Transplant.
Kidney Int. 71, 924–930 (2007). anti‑CD20 antibodies based on affinity constant and 13, 3103–3113 (2013).
126. Kalliakmani, P., Koutroulia, E., Sotsiou, F., epitope. Cancer Sci. 101, 201–209 (2010). 175. Alexander, T. et al. Depletion of autoreactive
Vlachojannis, J. G. & Goumenos, D. S. Benefit and cost 150. Tedder, T. F. & Engel, P. CD20: a regulator of cell-cycle immunologic memory followed by autologous
from the long-term use of cyclosporine‑A in idiopathic progression of B lymphocytes. Immunol. Today 15, hematopoietic stem cell transplantation in patients with
membranous nephropathy. Nephrology (Carlton) 15, 450–454 (1994). refractory SLE induces long-term remission through
762–767 (2010). 151. Grillo-Lopez, A. J. Rituximab (Rituxan/MabThera): the de novo generation of a juvenile and tolerant immune
127. Cravedi, P. Rituximab in membranous nephropathy: first decade (1993–2003). Expert Rev. Anticancer system. Blood 113, 214–223 (2009).
not all studies are created equal. Nephron 135, Ther. 3, 767–779 (2003). 176. Evaluate. Alexion reports presentation of membranous
46–50 (2017). 152. Salama, A. D. & Pusey, C. D. Drug insight: rituximab in nephritis clinical trials. http://www.evaluategroup.com/
128. Heijl, C. et al. Incidence of malignancy in patients renal disease and transplantation. Nat. Clin. Pract. Universal/View.aspx?type=Story&id=33783 (2002).
treated for antineutrophil cytoplasm antibody- Nephrol. 2, 221–230 (2006). 177. Ma, H., Sandor, D. G. & Beck, L. H. Jr. The role of
associated vasculitis: follow‑up data from European 153. Jain, P. & O’Brien, S. Anti‑CD20 monoclonal antibodies complement in membranous nephropathy.
Vasculitis Study Group clinical trials. Ann. Rheum. Dis. in chronic lymphocytic leukemia. Expert Opin. Biol. Semin. Nephrol. 33, 531–542 (2013).
70, 1415–1421 (2011). Ther. 13, 169–182 (2013). 178. Takano, T., Elimam, H. & Cybulsky, A. V. Complement-
129. US National Library of Medicine. ClinicalTrials.gov 154. van Imhoff, G. W. et al. Ofatumumab versus rituximab mediated cellular injury. Semin. Nephrol. 33, 586–601
https://clinicaltrials.gov/ct2/show/NCT03018535 salvage chemoimmunotherapy in relapsed or refractory (2013).
(2017). diffuse large B‑cell lymphoma: the ORCHARRD study. 179. Cattran, D. C. et al. Validation of a predictive model of
130. US National Library of Medicine. ClinicalTrials.gov J. Clin. Oncol. 35, 544–551 (2016). idiopathic membranous nephropathy: its clinical and
https://clinicaltrials.gov/ct2/show/NCT01180036 155. Cang, S., Mukhi, N., Wang, K. & Liu, D. Novel CD20 research implications. Kidney Int. 51, 901–907
(2017). monoclonal antibodies for lymphoma therapy. (1997).
131. US National Library of Medicine. ClinicalTrials.gov J. Hematol. Oncol. 5, 64 (2012).
https://clinicaltrials.gov/ct2/show/NCT01955187 156. Robak, T. & Robak, E. New anti‑CD20 monoclonal Acknowledgements
(2016). antibodies for the treatment of B‑cell lymphoid Luca Perico, Istituto di Ricerche Farmacologiche Mario Negri,
132. Fervenza, F. C. et al. A multicenter randomized malignancies. BioDrugs 25, 13–25 (2011). Bergamo, Italy, helped to revise the paragraph on PLA 2R
controlled trial of rituximab versus cyclosporine in the 157. Wierda, W. G. et al. Ofatumumab is active in patients epitope spreading and FIG. 2.
treatment of idiopathic membranous nephropathy with fludarabine-refractory CLL irrespective of prior
(MENTOR). Nephron 130, 159–168 (2015). rituximab: results from the phase 2 international study. Author contributions
133. Rojas-Rivera, J. et al. A European multicentre and Blood 118, 5126–5129 (2011). P.R. and F.C.F. researched data for the article. All authors
open-label controlled randomized trial to evaluate the 158. Taylor, P. C. et al. Ofatumumab, a fully human contributed to discussion of the article’s content, writing and
efficacy of sequential treatment with tacrolimus- anti‑CD20 monoclonal antibody, in biological-naive, review/editing the manuscript before submission.
rituximab versus steroids plus cyclophosphamide in rheumatoid arthritis patients with an inadequate
patients with primary membranous nephropathy: the response to methotrexate: a randomised, double-blind, Competing interests statement
STARMEN study. Clin. Kidney J. 8, 503–510 (2015). placebo-controlled clinical trial. Ann. Rheum. Dis. 70, F.C.F. has received unrestricted research grants from
134. Moroni, G. et al. Long-term outcome of renal 2119–2125 (2011). Genentech/Roche, the maker of rituximab. The other authors
transplantation in patients with idiopathic membranous 159. Basu, B. Ofatumumab for rituximab-resistant declare no competing interests.
glomerulonephritis (MN). Nephrol. Dial. Transplant. 25, nephrotic syndrome. N. Engl. J. Med. 370,
3408–3415 (2010). 1268–1270 (2014). Publisher’s note
135. Dabade, T. S., Grande, J. P., Norby, S. M., 160. Frieri, M., Heuser, W. & Bliss, J. Efficacy of novel Springer Nature remains neutral with regard to jurisdictional
Fervenza, F. C. & Cosio, F. G. Recurrent idiopathic monoclonal antibody belimumab in the treatment of claims in published maps and institutional affiliations.

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | SEPTEMBER 2017 | 579


©
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
.

You might also like