You are on page 1of 15

Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 337

Article
Primary Nephrotic Syndrome
Landan Zand, MD and Fernando C. Fervenza, MD, PhD, FASN
Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota

typically shows minimal or no changes on light microscopy, and


Learning Objectives immunofluorescence is either negative or shows scant immunoglob-
ulin deposition. The hallmark of the disease is presence of diffuse
1. To review recent evidence on the diagnosis and treatment of foot process effacement on electron microscopy (EM). In older
minimal change disease adults with heavy proteinuria, acute tubular necrosis is commonly
present on light microscopy (3). The majority of the cases of MCD
2. To determine the role of genetic testing in a patient with an are idiopathic (primary). In secondary forms, MCD has been associ-
FSGS lesion ated with drugs, neoplasms (primarily hematologic malignancies),
infections, and allergies (4). In this section we will focus on
3. To discuss updates on the significance of recently discovered
primary/idiopathic MCD.
antigens in the diagnosis and treatment of membranous
nephropathy Pathogenesis
4. To describe updates in the diagnosis and management of Both T and B cells have been suggested to play a role in the patho-
maladaptive FSGS genesis of MCD (5). The main proposed mechanism has been
secretion of permeability factor(s) that damages podocytes. This per-
5. To discuss the efficacy of treatment options in managing meability factor, however, has remained elusive until recently. Previ-
patients with membranous nephropathy ously suggested circulating factors included a vascular permeability
factor that mimics hemopexin (6–9), angiopoietin-like-4 secreted by
the podocyte (10), or cytokines such as IL-8 and IL-13 (11–13),
but none of these candidates were shown to be specific to patients
Introduction with MCD and were present in patients with NS from other
Nephrotic syndrome (NS) is defined as persistent proteinuria of diseases (14,15). More recently in a landmark paper, Watts et al.
$3.5 g/d (in adults) or $40 mg/h per m2 (in children) in combi- showed that nephrin autoantibodies are indeed the circulating factor
nation with hypoalbuminemia (,3.5 g/dl) (if measured by bromo- that results in diffuse podocytopathy in a subset of patients with
cresol green method) or ,3.0 g/dl (if measured by bromocresol MCD (16–18). In this study they evaluated the sera of 62 patients
purple or immunonephelometric methods) (1,2). Other features of with noncongenital and biopsy-proven MCD at the time of active
NS include edema (most commonly peripheral, but could present disease and showed that 18 of the 62 (29%) patients had circulating
with ascites or pleural effusion), hyperlipidemia, and a hypercoagul- nephrin autoantibodies. The authors showed that the nephrin auto-
able state. These additional features are not required to make a antibody levels either decreased significantly or were absent after
diagnosis of NS but are commonly present. Urine studies typically treatment and once patients entered partial remission or complete
show lipiduria, fatty casts, and oval fat bodies. Hypoalbuminemia remission, suggesting that indeed the nephrin autoantibodies play a
is essential for diagnosis of NS, and in the absence of hypoalbumi- role in the pathogenesis of the disease. The patients with positive
nemia the proteinuria is instead referred to as nephrotic-range nephrin antibodies were also noted to have evidence of fine punctate
proteinuria (1). This is a crucial differentiation because nephrotic- IgG staining on immunofluorescence and colocalization of IgG
range proteinuria in patients without edema or hypoalbuminemia is with nephrin on confocal microscopy. The authors proposed that it
more likely to be due to secondary FSGS (e.g., resulting from adap- is through this in situ binding of anti-nephrin IgG to nephrin that
tive changes such as glomerular hyperfiltration, obesity). NS arises the integrity of the slit diaphragm is disrupted and podocytopathy
as a result of glomerular disease primarily characterized by podocyte and proteinuria ensues.
damage, and it can be due to primary podocytopathies or it can be
secondary to conditions such as diabetes or amyloidosis. In this Therapies
section we will discuss the three main primary causes of NS, includ- Corticosteroids. Corticosteroids are historically considered first-line
ing minimal change disease (MCD), FSGS, and membranous therapy for treatment of MCD in both adult and children. The cor-
nephropathy (MN). ticosteroids commonly used include prednisone or prednisolone,
given as either daily dosing or alternate-day dosing. The optimal
duration of therapy with corticosteroid is not known. Several ran-
MCD domized trials in children suggest that a shorter duration of therapy
MCD is a primary podocytopathy that mainly affects children may be as effective as longer therapy without increasing the risk of
(90%) and less commonly adults (10%). A renal biopsy specimen relapse (19–21). However, a 2019 randomized controlled trial

Copyright # 2022 by the American Society of Nephrology nephsap.org


338 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

(RCT) of 237 children aged 1–14 years with steroid-sensitive NS comparing MMF with cyclosporine in children with frequently
comparing extended prednisolone treatment (16 weeks) with stan- relapsing NS showed similar results, with lower rates of remission
dard prednisolone treatment (8 weeks) found similar efficacy and and more relapses in MMF-treated patients (31). When compared
rates of relapse but reported that extended prednisolone treatment with levamisole in children with frequently relapsing NS, MMF
was more cost effective (owing to less primary care and hospital vis- showed equivalent efficacy, with similar rates of remission and
its) and was associated with better quality of life (21). This study, relapse (32). However, both treatments reduced exposure to corti-
however, was not able to assess long-term side effects associated with costeroids by 30%–45% (32).
corticosteroid therapy. There is less robust evidence available in Data for the use of MMF in adult patients with MCD is sparse.
adults. Traditionally, patients are treated for $3 months, with retro- A 2018 open-label RCT enrolled 119 adults with new-onset MCD
spective studies suggesting more patients enter remission with longer and compared high-dose prednisone (1 mg/kg per day) with low-
duration of therapy (22). In adults, alternate-day corticosteroid ther- dose prednisone (0.5 mg/kg per day) with enteric-coated mycophe-
apy shows no differences in remissions, time to remission, relapse nolate sodium (720 mg twice daily) as first-line therapy over a
rate, or time to relapse compared with daily-treated patients (23). 24-week period. The primary outcome was complete remission at 4
An RCT is required to evaluate short- versus long-term cortico- weeks, which was achieved in approximately 60% of patients in
steroid therapy in adults with MCD. Given the concerns regarding either arm. Both treatment arms had similar rates of complete remis-
complications related to a prolonged course of steroids in patients sion at 24 weeks, and rates of relapse and adverse events did not
with MCD, there has been a growing shift toward using steroid- differ between groups. This study suggests a potential role for myco-
sparing agents such as calcineurin inhibitors (CNIs) as first-line ther- phenolate to reduce exposure to corticosteroids (33).
apy, as discussed below.
Rituximab. Over the past two decades since the first report of
CNIs. Cyclosporine and tacrolimus have been used successfully in
successful treatment of a patient with steroid-dependent NS with
treating patients with steroid-dependent or frequently relapsing MCD
rituximab, there has been growing evidence from observational
in adults (24–27). The goal of CNIs is to minimize repeated expo-
studies and clinical trials (comparing rituximab with steroids or
sures to corticosteroids because relapses are common, with one-third
placebo) that rituximab is efficacious for the treatment of patients
of patients having more than one relapse (23). However, many
with steroid-dependent NS or frequently relapsing NS in children
patients will experience significant toxicity from the corticosteroids
(34–38). A large, randomized trial published in 2018 included
related to the first prolonged exposure, especially those with underly-
120 children with steroid-dependent NS, and compared rituximab
ing obesity or diabetes mellitus. As a result, newer trials have evaluated
(375 mg/m2 3 2) with prednisolone for 4 weeks versus tacrolimus
use of tacrolimus as first-line therapy. In an RCT of adult-onset
(0.2 mg/kg per day) with tapering alternate-day prednisolone,
MCD, 119 patients were treated with 10 days of intravenous methyl-
stopped by 6 months in those without relapse. In this study,
prednisolone, followed by treatment with tacrolimus or prednisone
a course of rituximab was more effective than tacrolimus in
for 16–20 weeks with taper over 18 weeks. Tacrolimus was noninfer-
maintaining remission, with relapse-free survival of 90% versus
ior to prednisone, with similar rates of remission and sustained remis-
sion and similar rates of relapse and time to relapse (28). Adverse 63% during 12 months of follow-up (odds ratio, 5.21; 95%
events and serious adverse events were more common in the predni- confidence interval [CI], 1.93 to 14.07). Median time to relapse
sone group (28). Another recent multicenter RCT from the United was 40 weeks in the rituximab-treated patients versus 29 weeks
Kingdom enrolled 50 patients with de novo MCD and compared a in the tacrolimus-treated group. In addition, mean cumulative
steroid-free protocol of tacrolimus with prednisolone therapy. Earlier corticosteroid dose was significantly lower in rituximab-treated
remissions were seen in prednisolone-treated patients, with a higher patients (39).
remission rate at 4 weeks and nonsignificantly greater remission rate at Several dosing regimens for rituximab have been used in chil-
8 weeks (84% in prednisolone group versus 68% in the tacrolimus dren, and two retrospective observational studies have reported on
group) but with similar rates of complete remission at 16 and 26 weeks outcomes with an immunosuppressive protocol. Lower-dose rituxi-
in both groups. Both groups had similar rates of relapses during the mab (100 mg/m2 or 375 mg/m2 3 1) in the absence of mainte-
follow-up period and very similar rates of adverse events (29). The nance immunosuppression has been associated with a higher rate of
overall number of patients in this trial was small and it is likely that relapse (hazard ratio for relapse, 1.55–4.98). Higher-dose rituximab
the study was underpowered to detect a difference between the two (375 mg/m2 3 2 or a cumulative dose of 750–1500 mg/m2)
groups. Taken together, these two trials suggest that tacrolimus may resulted in sustained remission, with relapse-free survival up to
be used as a first-line agent and certainly should be considered in 14 months in one series (40,41). However, those given lower-dose
patients who are at higher risk of complications from corticosteroids. rituximab with maintenance immunosuppression (defined as co-
treatment with steroids, CNIs, or mycophenolate) had similar
Mycophenolate Mofetil. Mycophenolate salts are available as either relapse-free survival to those treated with high-dose rituximab alone
mycophenolate mofetil (MMF) or enteric-coated mycophenolate (41). Prior studies in children with NS who are dependent on CNI
sodium. Limited data are available for their use in MCD. Thus far, and prednisone therapy have shown that the addition of rituximab
MMF has been used as a second- or third-line agent for the treatment allows for lowering of CNI and prednisone doses without increasing
of patients with frequently relapsing disease. In children with fre- rate of relapse, both short-term and long-term (42,43). However, a
quently relapsing NS, MMF was inferior to cyclosporine, with more prior RCT of 31 children with resistant NS demonstrated that addi-
relapses occurring in patients treated with MMF during the first tion of rituximab provides no additional benefit in children who are
year (but not the second year) (30). A more recent Chinese study resistant to prednisone and CNI therapy (44).
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 339

An ongoing trial is comparing rituximab with a newer anti- FSGS


CD20 agent (ofatumumab) in children with steroid-dependent NS FSGS, often incorrectly abbreviated as focal sclerosis, is a lesion
on stable doses of prednisone and CNI therapy (NCT02394119) identified on light microscopy with diverse causes and pathogenici-
(45). Fewer data are available in adults, but observational data and ties that are linked by podocyte injury and depletion (53). FSGS is
evidence from several small trials suggest that rituximab is similarly not a disease entity but rather a pattern of injury. It is not uncom-
effective in children, in maintaining remission and reducing the mon to find FSGS lesions in patients with other glomerulonephriti-
requirement for maintenance immunosuppression in both new- des such as lupus nephritis or ANCA-associated vasculitis, which
onset MCD and frequently relapsing NS (37,46–48). reflects postinflammatory scarring of a proliferative or necrotizing
lesion.
Adrenocorticotropic Hormone. Adrenocorticotropic hormone
In the absence of an underlying GN, FSGS is broadly catego-
(ACTH) was widely used in the 1950s for treatment of patients
rized into three groups to differentiate between the underlying path-
with NS. This was later replaced by corticosteroids. Over the past
ophysiologic changes that result in the formation of the segmental
decade, there has been renewed interest in ACTH on the basis of
scar. Primary (also known as idiopathic) FSGS is a lesion whose
case series and systemic reviews suggesting a potential role for
underlying cause has not yet been identified, but indirect evidence
ACTH in treatment of MCD despite scarce data (49,50). A recent
suggests a potential role for permeability factor(s) that cause diffuse
randomized trial compared 6 months of ACTH therapy with con-
podocyte damage. Secondary FSGS can be due to toxins or drugs
trol, with a crossover design (from control arm to ACTH arm in
(e.g., pamidronate) (54) or virus (e.g., HIV) (55), which result in dif-
case of relapse) in children with NS (51). The study was ended early
fuse podocyte damage. Secondary FSGS can also be the result of
because of the high rate of relapse in the ACTH arm (93%), sug-
hyperfiltration, either through structural adaptation of the glomeruli
gesting ACTH was ineffective in maintaining remission (51). There
to nephron loss (e.g., after partial nephrectomy) or through increased
are no randomized trials available in adults. At this point there are
demand on existing nephrons (e.g., obesity) (56–58). More recently,
not enough data to support use of ACTH in patients with MCD,
the class of genetic FSGS has been included under secondary FSGS
especially in children.
to reflect patients who have mutations either in podocyte genes or
glomerular basement membrane (GBM) collagen genes and who
Outcomes
present with lesions of FSGS. Still, despite extensive evaluation, a
Children with MCD typically have a good long-term outcome, and significant number of patients have a cause that is not identified and
progression to ESKD is rare. Steroid responsiveness is the best pre- these patients are classified as FSGS on undetermined cause (59,60).
dictor of outcome in these patients. The FSGS lesion is commonly Understanding the underlying pathophysiology is essential
identified on repeated biopsy specimens from patients who do not because it has significant implications on how to treat the patient.
respond to steroids. Even though long-term renal outcome is favor-
able in children with MCD, those with frequently relapsing disease Podocyte Injury and Death
requiring repeated courses of corticosteroids experience complica-
Podocytes are a key player in the pathogenesis of FSGS because they
tions related to the corticosteroids, including osteoporosis, obesity,
play a central role in maintaining the integrity of the filtration bar-
and hypertension.
rier. Damage to the podocyte is central to the development and pro-
In adults with MCD, the outcome is less favorable (52).
gression of disease. The initial podocyte injury leads to a variety of
Overall, 50% of patients respond to steroids and do not have
morphologic changes in the podocyte, the most prominent of which
recurrence of the disease, but close to 30% of patients have fre-
is foot process effacement (61). If injury is sustained or more severe,
quently relapsing disease. In addition, concomitant AKI related to
it results in podocyte apoptosis and detachment. For example, Yes-
acute tubular necrosis in adults is common and is associated with
associated protein has been shown to be an inhibitor of podocyte
increased time to remission after steroid therapy. Similarly, as in
apoptosis, and in animal models its suppression results in podocyte
children, response to immunosuppression is the best predictor of
apoptosis, proteinuric kidney disease, and the development of histo-
outcome in adults with MCD. Repeated biopsy in steroid-
logic features of FSGS (62,63). After podocyte loss, the denuded
unresponsive patients may be indicated, and not uncommonly an
GBM comes into contact with the parietal epithelial cell, resulting
FSGS lesion is identified. Genetic testing should be considered in in scar formation (64). Parietal epithelial cells may act as a progeni-
patients who do not respond to steroids, and this is discussed in tor and replace the lost podocytes (65). There may, however, be a
greater detail below. critical threshold of podocyte depletion, after which repair may not
be possible and progression of the disease may be inevitable (66).
A variety of different mechanisms can result in podocyte injury
4 Circulating nephrin autoantibody is the cause of and death. In primary FSGS, a soluble factor(s) results in diffuse
podocytopathy in about 30% of the patients with podocyte damage. Proposed candidates have included soluble
MCD and the antibody titers decrease significantly urokinase-type plasminogen activator receptor, cardiotrophin-like
as patients enter remission. cytokine-1, and CD40 antibodies, but none have proved to be spe-
cific (67–70). In secondary FSGS, the mismatch between glomerular
4 One hypothesis is that in situ binding of anti- demand and glomerular capacity results in glomerular hypertension
nephrin IgG disrupts the integrity of the slit and increased fluid shear stress on the podocytes, which in turn
diaphragm causing the podocytopathy. results in podocyte loss and development of segmental scars (58).
Genetic forms of FSGS (familial or sporadic) result from mutations
340 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

in genes encoding proteins that are essential for maintenance of Treatment. The goal of therapy in patients with primary FSGS is to
podocyte cytoskeleton, foot processes, slit diaphragm, or interaction induce remission (either complete or partial) as it has been associated
between the podocyte and the basement membrane (71). with improved long-term outcome. Historically, the majority of the
drug trials in primary FSGS have suffered from the fundamental flaw
Importance of Renal Biopsy in Classification of FSGS of enrolling a heterogeneous group of patients comprising not just
FSGS is diagnosed when at least one glomerulus in a biopsy sample patients with primary FSGS but also those with secondary or genetic
shows the typical appearance of a segmental increase in glomerular causes (59,80). This has resulted in many negative trials and inability
matrix with obliteration of glomerular capillaries and scar formation to interpret the data. Despite these limitations, there is no doubt that
(72). Thorough evaluation of a biopsy sample by light microscopy in patients with presumed primary FSGS, immunosuppression is
and EM is required to accurately classify primary versus secondary required. In general, angiotensin-converting enzyme inhibitors and
FSGS, and exclude other causes of an FSGS lesion (73). A classi- angiotensin receptor blockers are recommended, but alone they are
fication system on the basis of light microscopic appearance has inadequate to treat patients with primary FSGS.
been proposed, whereby five morphologic patterns have been identi- Corticosteroids. Historically, corticosteroids have been the first-
fied: FSGS not otherwise specified, tip, perihilar, cellular, and col- line therapy for patients with primary FSGS. Prolonged courses
lapsing variants (72,74,75). However, light microscopic appearance ($16 weeks) of high-dose steroids (1 mg/kg) may be required to
alone cannot differentiate between primary, secondary, and genetic achieve remission. Up to 30% of patients will have a relapse after
FSGS (74). tapering or stopping corticosteroids, and a second course of steroids
An important consideration in the evaluation of a kidney may not be as effective (81). If patients do not respond to steroids
biopsy sample showing FSGS, in any of its light microscopic var- after 16 weeks of therapy, second-line therapy such as CNIs should
iants, is the degree of foot process effacement on EM examination be considered. A recent study suggests that proteinuria response at 8
(53). The degree of foot process effacement is determined primarily weeks is an excellent predictor of the overall response to immuno-
by the nature of the pathogenic process affecting the podocyte (76). suppressive therapy (82). This was a retrospective cohort study
Patients with primary FSGS show diffuse foot process effacement, including 51 patients (mean age 47 years) treated with corticoster-
oids for a median of 17 weeks. Ten patients achieved a partial
whereas foot processes are relatively preserved in cases of secondary
response at 8 weeks with a total of 18% and 35% achieving com-
FSGS, with little overlap between the two (77). Thus, the degree of
plete response and partial response, respectively. A decrease of
foot process effacement by EM is a crucial clue to distinguishing pri-
.20% predicted remission.
mary from secondary forms of FSGS, with some exceptions, such as
CNIs. CNIs are typically used as second-line therapy for treat-
cases of “collapsing” FSGS secondary to HIV, IFN, or pamidronate
ment of patients who are resistant to corticosteroids (83,84). CNIs
therapy that are characterized by widespread foot process effacement
should also be considered as first-line therapy in patients who have
on EM. However, appearance on light microscopy or EM cannot
contraindications to using corticosteroids, such as those with obesity,
reliably distinguish a genetic cause from a primary form of FSGS
diabetes mellitus, or osteoporosis (85). The effectiveness of CNIs is
(see below).
partly due to immunosuppressive effects but may also partly relate
to their stabilization of the actin cytoskeleton in podocytes (86).
Primary FSGS
Nephrotoxicity related to CNIs remains a concern, especially in
Primary, or idiopathic, FSGS is a primary podocytopathy that those with impaired renal function (84,87,88). Voclosporin, a new
results from circulating factor(s) that cause diffuse podocyte damage. CNI agent, was under investigation as first-line therapy in treating
The underlying pathophysiology resembles that of MCD, and patients with primary FSGS but the trial was ended early because of
although MCD and primary FSGS are commonly considered to be difficulty with recruitment.
two separate diseases, some have proposed that they are different his- Anti-CD20 Therapy. Rituximab has been shown to be effective
tologic manifestations of the same disease process (78). Inasmuch as in the treatment of children and adults with FSGS who are
the disease is the result of diffuse podocyte damage, the onset tends steroid responsive but remain steroid dependent or who experience
to be sudden, and patients present clinically with NS ($3.5 g/d of frequent relapse (37,48,88). Addition of rituximab to existing
proteinuria in association with hypoalbuminemia ,3.5 g/dl). Histo- immunosuppression has been associated with increased rates of
logically, light microscopy reveals FSGS lesions but, more impor- remission, reductions in relapse rates, and reduced need for addi-
tantly, EM shows diffuse foot process effacement (.80%) (74,79). tional immunosuppression, in retrospective and observational stud-
It is important to note that the diagnosis of presumed primary ies (39,46,87,89). Its use in patients with steroid-resistant FSGS is
FSGS in not a pathologic diagnosis but a clinical-pathologic one, less clear (89–91).
necessitating careful evaluation of the clinical, laboratory, and kidney Newer anti-CD20 agents such as obinutuzumab have been
biopsy findings (80). It is a diagnosis of exclusion, and distinction shown to be effective in the treatment of patients with other paren-
between primary and secondary FSGS is not always obvious. Failure chymal diseases such as lupus nephritis and those with MN resis-
to accurately identify the cause of FSGS can result in patients with tant to rituximab therapy (92,93). Its use in treatment of patients
secondary or genetic forms of FSGS undergoing unnecessary immu- with treatment-dependent or -resistant primary FSGS is currently
nosuppressive therapy. Until a specific biomarker is identified, a under investigation in a prospective single arm clinical trial
detailed and comprehensive evaluation of the native kidney biopsy (NCT04983888).
specimen, including EM examination, together with the clinical and Other. Data regarding use of MMF in patients with primary
laboratory findings, provides the best chance of approaching the cor- FSGS are less robust. MMF is likely inferior to CNI therapy but
rect diagnosis. may be used in combination with low-dose steroids for treatment of
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 341

patients who have impaired renal function and are unable to take The type of FSGS was not clarified in this study but the median
CNIs (94). In very small case series, ACTH has been shown to albumin-creatinine ratio in the cohort was 1248 (range, 749–2211)
lower proteinuria in some patients with steroid-resistant FSGS mg/g, suggesting that the majority of patients either had secondary
(95,96). A clinical trial evaluating the role of ACTH in treatment- or genetic FSGS (99). The primary outcome was composite of
resistant FSGS (NCT02633046) has been completed but results on sustained eGFR decline of 40% or death because of kidney or
the efficacy of the drug have not yet been published. cardiovascular causes. There was higher rate of primary outcome in
Sparsentan is a combination agent, comprising an endothelin placebo (21%) compared with dapagliflozin (11.3%) but this did
type A receptor antagonist and the angiotensin receptor blocker not reach statistical significance (hazard ratio, 0.46; 95% CI, 0.17 to
irbesartan. Its use was studied in a phase 2 trial of sparsentan dose 1.24) likely because of the small number of patients in this subgroup
escalation in patients with primary FSGS, where the primary end of patients. Additional studies in larger cohorts are needed to evalu-
point was change in proteinuria from baseline to week 8 (97). ate the efficacy of SGLT-2 inhibitors in this population.
Endothelin type A receptor antagonists have been shown to reduce
proteinuria in rodent models of FSGS, and they may augment the Genetic FSGS
effect of inhibitors of the renin-angiotensin system. The study Genetic FSGS refers to monogenic forms of FSGS in which genes
enrolled 109 patients across three dosing arms (36 treated with encoding proteins expressed on either the podocyte or the GBM
irbesartan and 73 treated with varying doses of sparsentan). Signifi- are mutated. The clinical presentation of patients with genetic
cantly greater reductions in proteinuria were observed in pooled FSGS can vary widely, which makes it difficult for clinicians to
sparsentan-treated versus irbesartan-treated patients (244.8%; 95% select the patients who would benefit from testing. Patients can
CI, 252.7% to 235.7% versus 218.5%; 95% CI, 235.6% to present with NS, nephrotic-range proteinuria, or subnephrotic pro-
1.7%). However, it should be pointed out that even though the teinuria, and the degree of foot process effacement on EM can be
study aimed to enroll patients with primary FSGS, the inclusion variable (71,72). However, full-blown NS in adults with genetic
criteria were such that patients with secondary and genetic forms of forms of FSGS is uncommon. In some cases, extrarenal features
FSGS were also included. The main benefit was seen in patients may be present, and the FSGS may be part of a syndromic disease,
without NS; therefore, the results may not apply to patients with but in the majority of the patients extrarenal manifestations are
primary FSGS. uncommon. Genetic FSGS more commonly affects the pediatric
population, but up to 14% of adult cases are due to genetic muta-
Secondary FSGS tions (100). Several retrospective studies have identified significant
Adaptive secondary FSGS is due to glomerular capillary hyperten- rates of gene mutations (#30%) in patients with steroid-resistant
sion and hyperfiltration as a result of mismatch between glomerular NS despite clinical presentation and histologic findings that mimic
load and glomerular capacity. Patients with secondary FSGS (in the primary FSGS (101–103). Conversely, genetic testing in a small
absence of a virus or drug) present less acutely and without features subgroup of patients with steroid-sensitive NS failed to reveal any
of NS. The proteinuria may be nephrotic or subnephrotic but is gene mutations (100). Therefore, adults who appear to have pri-
typically associated with a normal serum albumin (.3.5 g/dl). His- mary FSGS but do not respond to therapy should be tested for a
tologically, light microscopy shows an FSGS lesion, but EM reveals genetic cause of FSGS. Other groups that may benefit from genetic
mild-to-moderate foot process effacement (,80%) (72,74). testing are those who appear to have secondary FSGS in the
absence of an obvious cause (e.g., no evidence of reduced nephron
Treatment. Treatment is focused on mitigating factors that increase mass or obesity) and those who cannot be easily categorized into
glomerular demand, including weight loss, tight BP control, and primary or secondary FSGS (undetermined FSGS) when there is a
treatment of sleep apnea. Renin-angiotensin-aldosterone system mismatch between their clinical presentations and the degree of
(RAAS) blockade with either an angiotensin-converting enzyme foot process effacement on biopsy (72,104) (Figure 1). In a recent
inhibitor or an angiotensin receptor blocker is strongly recom- prospective study of 49 consecutive patients with an FSGS lesion
mended to lower proteinuria and control BP. The protective effect who underwent genetic testing, patients with undetermined FSGS
of RAAS blockade is partly mediated by vasodilation of efferent had the highest rate of positivity (87.5%, 7 of 8) followed by
arterioles and lowering intraglomerular hypertension. More recently, patients with secondary FSGS without an identifiable cause
interest has grown in evaluating the role of sodium-glucose cotran- (61.5%, 8 of 13). Genetic testing may also be indicated in patients
sporter 2 (SGLT-2) inhibitors in further lowering glomerular hyper- younger than 18 years of age, those with a family history of FSGS,
tension. Their proposed mechanism of action is via increased and those with syndromic forms of FSGS.
sodium delivery to the macula densa, which, through tubuloglomer- Traditionally, testing for a few of the most common mutations
ular feedback, results in afferent arteriolar vasoconstriction and was carried out by Sanger sequencing. However, with the growing
reduction in intraglomerular hypertension. They have been shown recognition of genes that can cause genetic FSGS (now .50 genes)
to reduce proteinuria and delay progression of kidney disease in the preferred method has moved to next-generation sequencing,
both diabetic and nondiabetic patients (75,98). The DAPA-CKD which allows sequencing of a large number of genes. Whole exome
trial randomized 4304 patients with eGFR between 25 and 75 sequencing is a technique in which the exome (part of the DNA
ml/min per 1.73 m2 and albumin-creatinine ratio between 200 and that encodes proteins) is sequenced with the goal of finding patho-
5000 mg/g to receiving dapagliflozin 10 mg daily versus placebo. genic mutations. This method has been successfully used to identify
Subgroup analysis was performed on 104 patients with biopsy- mutations in patients with FSGS and CKD of unknown cause
proven FSGS (45 receiving dapagliflozin and 59 receiving placebo). (102,103,105,106).
342 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

To date, the majority of genes that have been associated with forms with secondary forms seen in association with other diseases
genetic FSGS are those encoding proteins involved in the mainte- such as sarcoidosis, malignancy, systemic autoimmune diseases
nance of podocyte cytoskeleton, slit diaphragm, and foot processes. such as SLE, hepatitis B, and drugs. There is now growing recog-
There is increasing evidence that patients with FSGS lesions com- nition that both primary and secondary MN are due to circulating
monly have mutations in genes encoding type IV collagen autoantibodies against specific antigens on the podocyte surface or
(COL4A3, COL4A4, COL4A5) in the absence of any evidence of associated with overexpression of a specific antigen (reviewed in
abnormality in the GBM (105–109). In a recent study, 193 indivi- Hoxha et al.) (111). Therefore, a target antigen-based approach
duals who were identified either by having biopsy-proven FSGS or has been proposed for the classification of MN (112) rather than
by the presence of proteinuria and a family member with FSGS categorizing the disease as primary versus secondary. Antigens that
were examined for abnormalities in 109 genes associated with are more commonly seen in the absence of an associated disease
FSGS, GBM abnormalities, and congenital abnormalities of the kid- include M-type phospholipase A2 receptor 1 (PLA2R), thrombos-
neys and urinary tract. Genetic abnormalities were identified in pondin type 1 domain-containing 7A (THSD7A), neural EGF-
11% of cases, of which 55% were identified in genes encoding type like 1 protein (NELL-1), semaphorin 3B (Sema3B), protocadherin
IV collagen, and 40% in podocyte genes. In this series, abnormali- PCDH7 (PCDH7), and serine protease high-temperature require-
ties of COL4A3, COL4A4, and COL4A5 were the commonest ment A1 (HTRA1) (113–118). There are likely other antigens on
monogenetic cause of FSGS in adults (107). Therefore, these genes the podocyte surface with associated autoantibodies that cause
should be considered for testing in evaluating a patient for genetic MN that are yet to be identified. Other antigens seen commonly
FSGS. in association with other diseases including lupus and post bone
There are currently no specific treatments available for marrow transplantation include exostosin 1 and 2 (EXT1/EXT2)
patients with genetic forms of FSGS. Response to immunosup- (119), neural cell adhesion molecule 1 (NCAM-1) (120,121), and
pression is suboptimal and should be avoided (110). Focus should protocadherin FAT1 (FAT1).
be on conservative measures, including weight and BP control in
addition to use of RAAS blockade if BP allows. SGLT-2 inhibitors Anti-PLA2R Serology for Diagnosis, Prognosis,
may also provide benefit in these patient but more studies are
needed.
and Management
The diagnosis of MN is on the basis of the renal biopsy findings of
an MN lesion with the associated typical immune complex deposits.
MN PLA2R-associated MN can be diagnosed for a renal biopsy specimen
MN is a pattern of injury the hallmark of which is the presence of showing staining of the PLA2R antigen in the deposits. This can be
immune complexes in the subepithelial region of the glomeruli. done either by immunofluorescence or by immunoperoxidase stain-
Traditionally, MN has been divided into primary and secondary ing on paraffin-embedded kidney tissue that has undergone pronase

Figure 1. Opinion-based approach to genetic testing in adult-onset FSGS. Note that viral- and drug-associated forms of FSGS are usually
excluded by clinical and serologic evaluation. EM, electron microscopy; FPE, foot process effacement. Reprinted from ref. 72, with permission.
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 343

digestion (122). In cases of PLA2R-associated MN, the staining is PLA2R1 is a transmembrane receptor that has a large extracel-
intense along the glomerular capillary walls, whereas in negative cases lular domain with three distinct regions, including a cysteine-rich
the staining is either absent or faint (minimal “blush”) (122). Given domain (CysR), a fibronectin type II domain (FNII), and eight dis-
the specificity of anti-PLA2R antibodies in patients with MN, the tinct C-type lectin domains (CTLD1–8) (137). The most com-
data suggest that a kidney biopsy could be safely deferred in patients monly recognized epitope by the PLA2R antibody is the CysR
with positive anti-PLA2R antibodies who have preserved renal func- region, but over time the antibodies may recognize other epitopes,
tion (eGFR .60 ml/min) and no evidence of secondary MN or dia- including CTLD1 and CTLD7. The phenomenon whereby new
betes. (123). These results were further confirmed in a validation epitopes (typically within the same molecule) are recognized by the
cohort in which 101 patients with biopsy-proven MN and positive immune system is known as epitope spreading. In patients with
PLA2R antibodies were evaluated. Of the 79 patients with an eGFR PLA2R-associated MN, epitope spreading was suggested to portend
$60 ml/min per 1.73 m2, addition of kidney biopsy did not change a poor prognosis (137,138). However, in a prospective study of
the diagnosis or the management in any of the patients (124). 150 patients with newly diagnosed PLA2R-associated MN, the
PLA2R antibody has been shown to be highly specific for the authors elegantly showed that detection of a specific epitope was
diagnosis of MN and has not been detected in other glomerular dis- predominantly dependent on the PLA2R antibody level, and it was
eases or healthy control individuals. However, there are a handful of the antibody level (rather than the epitope recognition) that pre-
reports of patients with secondary MN who have positive PLA2R anti- dicted renal outcome and response to therapy (139). In other words,
bodies. It is possible that some of these cases are not true secondary it was the PLA2R antibody level that determined how many
but rather represent patients with PLA2R-associated MN who happen additional epitopes on the PLA2R antigen it would bind to; by
to have a secondary disease as well. This is supported by reports of diluting the PLA2R antibody titers, the antibody was no longer
cases that did not enter remission despite treatment of the secondary able to recognize the less common epitopes such as CTLD1 or
cause or cases that entered spontaneous remission without treatment CTLD7. These authors also identified another epitope (CTLD8),
of the secondary cause (125). Patients with secondary MN who most which had been previously described, that the antibody could
commonly have been found to have positive PLA2R antibodies recognize (139).
include those with hepatitis B, hepatitis C, and sarcoidosis (126–128). Monitoring PLA2R antibody levels over time is of great value
In a study of 39 patients with hepatitis B–associated MN, 25 patients when treating patients with PLA2R-associated MN. A decline in
had positive PLA2R staining on kidney biopsy specimens (128). The PLA2R antibody level over time is highly predictive that the patient
authors showed that the hepatitis B antigen colocalized along the glo- will enter remission (140–142). Therefore, a serology-based approach
merular capillary loop, which suggests that hepatitis B may trigger when treating patients with PLA2R-associated MN has been sug-
an immune response to PLA2R antigen in these patients (128). gested (Figure 2) (143). In patients treated with immunosuppression
The mechanism by which this may occur remains unknown. who show a rapid decline in PLA2R antibody within the first
A variety of different assays have been developed over the past
6 months, and the antibody eventually disappears, immunosuppres-
decade to detect anti-PLA2R antibodies. The ELISA (Euroimmun,
sion could be safely discontinued. On the other hand, if the patient
L€ubeck, Germany) is the most commonly used commercial assay
does not show any appreciable decline in PLA2R antibody within the
that allows quantitative assessment of the anti-PLA2R antibody.
first 6 months, a different immunosuppressive agent should be con-
However, it is not as sensitive as other assays. The cell-based indirect
sidered (143). Disappearance of PLA2R antibody (known as immu-
immunofluorescence assay (Euroimmun) is sensitive and may be use-
nologic remission) precedes improvement in proteinuria (clinical
ful in detecting low levels of anti-PLA2R antibodies, but it does not
remission). In patients who have reached immunologic remission but
allow for quantitative assessment, and its use is not as widespread.
continue to have proteinuria, additional immunosuppression is not
Some investigators suggest that both should be used simultaneously
for the initial diagnosis (123). Addressable laser bead immunoassay recommended but rather patience to allow for repair and remodeling
(Mitogen Advanced Diagnostics Laboratory, Calgary, Canada) is the of the basement membrane to occur. In these patients the use of a
most sensitive and quantitative assay available, but it is not commer- SLGT-2 inhibitor can be considered.
cially in use (129).
Antibody titers have been helpful in predicting patients who Other Serology
would enter spontaneous remission and those who would respond to THSD7A. THSD7A is another target antigen on the podocyte sur-
therapy. Low levels of antibody titer have been associated with high face that is responsible for the development of MN. Antibodies to
rates of spontaneous remission (130,131), whereas high levels are asso- THSD7A are present in 10% of MN patients who are PLA2R nega-
ciated with progressive loss of renal function (131–133). The associa- tive, accounting for 3% of cases of MN overall (114). The PLA2R
tion between baseline PLA2R antibody titers and proteinuria is less antibody and the THSD7A antibody were originally thought to be
clear, and this may be due to the lag between detection of antibodies mutually exclusive; however, since the publication of the original
in the blood and the development of proteinuria. Indeed, in a large study, a handful of cases with dual positivity have been reported
retrospective study using Department of Defense repository samples, (144). The method of antibody detection in original reports was
anti-PLA2R antibodies were detected at a median of 274 days (inter- Western blot, which is not commercially feasible. An indirect immu-
quartile range, 71–821 days) before a diagnosis of MN on the basis of nofluorescence assay has been developed but is not as commonly
renal biopsy (134). In a subset of these patients, PLA2R antibodies available commercially. Similar to PLA2R antibody, THSD7A anti-
were also found to predate the onset of nonnephrotic-range protein- body, detected by ELISA (Euroimmun), is helpful in both the diag-
uria. Declining levels of PLA2R over time are shown to be the best nosis and the treatment of patients, and antibody titers over time
predictor of response and have the best prognostic value (135,136). could be used to monitor disease activity (143). A notable difference
344 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

Figure 2. Prognostic algorithm. Although the temporal relationship between PLA2R antibody (Ab) levels and disease activity is well established,
a time lag of several months between immunologic and clinical response should be taken into account. Measurement of PLA2R Ab may obviate
the need for a “wait and see” period of 6 months, as recommended by the Kidney Disease: Improving Global Outcomes guidelines, and allow
more rapid treatment decisions. Serial PLA2R Ab measurements may guide the decision to start immunosuppression (IS). Patients with initial
high PLA2R Ab titers should be followed monthly, whereas those with moderate to low PLA2R Abs should be re-evaluated every two months.
This recommendation does not apply to patients with rapidly declining renal function, in whom prompt initiation of IS may be warranted.
PLA2R Ab ELISA (Euroimmun): low, 14–86 U/ml; moderate, 87–204 U/ml; and high, $204 U/ml. SCreat, serum creatinine. Reprinted
from ref. 143, with permission.

between PLA2R-positive and THSD7A-positive patients with MN malignancy. As such, patients who are positive for anti–NELL-1 anti-
is the high rate of malignancy in patients with THSD7A positivity. bodies in the biopsy specimen need to be evaluated for underly-
In the largest series of THSD7A cases, eight of the 40 patients were ing malignancy.
found to have an underlying malignancy within 3 months of diagnosis
of MN (145). Therefore, in patients with THSD7A-associated MN, a Sema3B. Sema3B was identified in 11 cases of PLA2R-negative
search for an underlying malignancy is highly recommended. MN. Sema3B localized as granular deposits along the GBM by
immunohistochemistry (116). In four of 11 cases, kidney biopsy
NELL-1. NELL-1 is one of the two antigens most recently recog- specimens also showed tubular basement membrane deposits of
nized as a cause of MN (115). NELL-1 was identified through laser IgG. In five patients, Western blot analysis showed reactivity to
microdissection of the glomeruli, followed by mass spectrometry. In reduced Sema3B in four of four patients with active disease and no
evaluating a total of 210 PLA2R-negative MN patients, 34 (16%) reactivity in one patient in clinical remission. Eight (73%) of the
were found to be NELL-1 positive (115). Therefore, NELL- 11 cases of Sema3B–associated MN were pediatric cases, and in five
1–associated MN is the second most common form of primary patients the disease started at or below the age of 2 years. This
MN. In this cohort, immunohistochemistry was also performed, suggests that Sema3B–associated MN appears to be a distinct type
which confirmed NELL-1 positivity. Patients with NELL-1– of MN more commonly seen in pediatric patients. Sema3B–asso-
associated MN are older (average age, 63 years), with no sex predilec- ciated MN has been shown to recur post kidney transplantation in
tion. The most common IgG identified was IgG1, but IgG2, IgG3, association with positive anti–Sema3B antibodies confirming that
and IgG4 were also present in some cases. This is in contrast to anti–Sema3B plays a role in the pathogenesis of this disease and
patients with PLA2R-associated MN, in whom the IgG is typically levels should be monitored peritransplantation (146).
IgG4. Anti–NELL-1 antibody was identified in a subset of these
patients, and in patients in whom longitudinal sera was available, a PCDH7. PCDH7 was detected via mass spectrometry in ten patients
reduction in NELL-1 antibody preceded clinical remission. It is there- out of 175 patients with negative PLA2R, THSD7A, NELL-1,
fore likely that the concept of immunologic remission and serial Sema3B, and EXT1/EXT2 (5.7%) from the Mayo Clinic cohort.
monitoring of antibody levels also applies to patients with NELL- Majority of patients were men (70%) with an average age of
1–associated MN. Antibody testing for NELL-1, however, is currently 61.2610.4 years with baseline creatinine of 1.3 mg/dl and 24-hour
not available commercially. It should also be noted that of the urinary protein of 4.2 g/24 h. One notable finding of the kidney
34 patients with NELL-1–associated MN, four (12%) had underlying biopsy in these patients was the absence of C3 staining in six of ten
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 345

patients and only 11 staining in the remaining four patients, suggest- Similar to EXT1/EXT2, patients were predominantly female (70%)
ing lack of complement activation in this subtype of MN. The with an average age of 34612 years with baseline creatinine of
IgG subtype was a combination of IgG1 through 4. An additional 1.3560.88 mg/dl with an average proteinuria of 6.769.5 g/24 h and
four patients were identified from the Belgian and French cohort serum albumin of 2.260.78 g/dl. Twenty-five percent of patients had
and taken together the prevalence of PCDH7-associated MN is concomitant proliferative features associated with class III and IV
estimated to be about 1.6%–2% of the biopsies with MN. Of the lupus nephritis. Anti–NCAM-1 antibody was identified in two
14 patients, six patients had associated autoimmune disease, sar- patients with available sera and was absent in patients with
coidosis, and underlying malignancy (117). Sera of six patients EXT1/EXT2 MN, suggesting presence of anti–NCAM-1 antibodies
with PCDH7-associated MN showed presence of anti-PCDH7 in a subset of lupus patients.
autoantibodies.
FAT1. MN can be a complication in patients who undergo hemato-
EXT1/EXT2. EXT1/EXT2 are another set of novel antigens that poietic stem cell transplantation (HSCT) and develop graft versus
have been identified in patients with MN (119). Similar to NELL-1, host disease. The antigen in this subset of patients has remained
the antigens were initially identified on mass spectrometry, and immu- unknown until recently when FAT1 was identified to be the puta-
nohistochemistry later confirmed staining for EXT1/EXT2. Of the tive antigen (120). In this study, a total of 14 patients with FAT1-
224 PLA2R-negative cases of MN, 26 (11.6%) had staining for associated MN were identified, all of whom had undergone HSCT.
EXT1/EXT2 by immunohistochemistry. These patients are younger MN occurred 2.460.8 years after HSCT, predominantly in men
(mean age, 35.7 years) and are predominantly female (80.8%). They (64%), with mean serum creatinine of 1.460.5 mg/dl and protein-
typically have preserved renal function, with a mean creatinine of 1.0 uria of 7.6 g/24 h at time of diagnosis. FAT1 antibody was identi-
mg/dl and average proteinuria of 5.9 g/24 h. In addition, 70% of the fied in serum of one patient who underwent testing.
patients had evidence of an underlying autoimmune disease with posi-
tive serology. Indeed, 34.6% of the patients had an underlying diagno- Treatment
sis of SLE. When biopsy specimens that were consistent with class V
All patients with MN should initially be given supportive therapy
lupus nephritis were specifically evaluated, eight out of 18 (44%) had
aiming to reduce proteinuria. This should include dietary sodium
positive staining for EXT1/EXT2. In the validation cohort in this
restriction to ,2 g/d, low-protein diet (0.8–1 g/kg per day), and BP
study, in the evaluation of patients who were PLA2R and THSD7A
control aiming at a systolic BP ,130 mm Hg if tolerated.
negative without evidence of class V lupus nephritis, three of 16
Since the recognition of PLA2R as the primary antigen in pri-
patients had staining for EXT1/EXT2. However, on further review all
mary MN and the advent of serial PLA2R antibody measurement,
three patients had evidence of underlying autoimmune disease, and
the approach to treating patients with PLA2R-associated MN has
two patients later received diagnoses of SLE. In addition, many of the
evolved over time. The goal of therapy now is to induce immuno-
biopsy specimens had features of secondary MN on histologic analysis,
logic remission, which corresponds to disappearance of the PLA2R
including staining for C1q, mesangial deposits, and subendothelial
antibody (60). Clinical remission (corresponding to improvement
deposits in addition to tubuloreticular inclusions. Therefore,
in proteinuria) typically follows 6 to 24 months after immuno-
EXT1/EXT2 are proteins that are overexpressed in the secondary form
of MN in association with underlying autoimmune disease, most com- logic remission has been achieved (149). In patients who do not
monly lupus. In this study, antibodies to EXT1/EXT2 could not be have antibodies detectable at the time of diagnosis, the traditional
identified. The absence of antibodies to EXT1/EXT2 in this study approach of monitoring serum albumin and urinary protein is
does not rule out the possibility that such an antibody exists, but it also recommended.
raises the possibility that EXT1/EXT2 may not be the target antigens Before deciding whether or not to treat a patient with MN, it is
themselves. Additional studies are required to further elucidate this. important to risk stratify because many patients may enter spontane-
In a more recent study of the 374 biopsies with membranous lupus ous remission without the need for immunosuppressive therapy.
nephritis, 122 (32.6%) were positive for EXT1/EXT2 and the rest were The risk of progression should be determined on the basis of clinical
negative. Patients with EXT1/EXT2-positive lupus MN were younger and immunologic parameters. Patients are considered to be at low
with lower serum creatinine, higher proteinuria, and less chronicity on risk of progression if they have normal or stable eGFR over the pre-
biopsy and had much lower rate of progression to ESKD compared ceding 6 months with proteinuria ,4 g/24 h with either negative
with those with EXT1/EXT2 negative membranous lupus nephritis PLA2R or low PLA2R antibody levels (,50 RU/ml) that are decreas-
(147). These data show that patients with EXT1/EXT2 have favorable ing by $25%. These patients do not require immunosuppressive
prognosis and perhaps a period of monitoring should be highly consid- therapy and should be treated conservatively with good BP control,
ered in this population before proceeding with immunosuppressive RAAS blockade, statin therapy, and modification of dietary protein
therapy. A recent study in 165 patients with class V lupus nephritis con- intake. Renal function, serum albumin, and 24-hour proteinuria
firmed that patients who were positive for EXT1/EXT2 had lower should be monitored every 6 months. Patients at moderate risk of
scores for activity index and lower chronicity tubular and atrophy scores progression are those who have normal or stable eGFR over the
compared with patients that were EXT1/EXT2 negative (148). preceding 6 months; however, proteinuria is between 4 and 8 g/24 h
(in the absence of life-threatening NS; e.g., complications such as a
NCAM-1. NCAM-1 is another antigen that is found to be overex- thromboembolic event), with moderately elevated PLA2R levels
pressed in patients with membranous lupus nephritis (121). Of 212 (.50 RU/ml but ,150 RU/ml) that are stable over a span of
patients with membranous lupus 14 patients (6.6%) were NCAM-1 6 months. These patients may need to be treated depending on how
positive. In this same cohort 15.8% were EXT1/EXT2 positive. they progress over time. For example, if the proteinuria increases,
346 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

eGFR declines, or PLA2R titer rises, then the patients should be trea- 6 or oral tacrolimus for 6 months (and then taper over 3 months)
ted. Options include rituximab or cyclic cyclophosphamide plus glu- and 1 g of rituximab on month 6. The primary outcome was rate of
cocorticoids. Alternative treatment includes CNIs (discussed below). complete or partial remission at 24 months. In the cyclophosphamide
Patients who are at high risk of progression should be treated without group, 60% achieved complete remission at 24 months versus 26% in
any additional observation period. These patients include those the tacrolimus/rituximab group (relative risk, 2.36; 95% CI, 1.34 to
who have a decline in their eGFR $25% related to the MN, have 4.16). Patients also achieved remission faster in the cyclophosphamide
.8 g/24 h of proteinuria, have life-threatening NS, or have PLA2R group. This study, however, is not a direct comparison of rituximab
antibodies that are elevated ($150 RU/ml) and are not declining with cyclophosphamide as patients were given rituximab 6 months
.25% within a 1- to 2-month interval. These patients require into the trial while receiving a CNI which is known to be inferior
immunosuppressive therapy. Recent trials suggest that rituximab may to rituximab and subsequently received a low dose of rituximab (only
be equally effective, with more remission durability (discussed below). 1 g) with no redosing at 6 months, and no information was available
Historically, alkylating agents (e.g., cyclophosphamide or chlor- as to whether patients were adequately B cell depleted. In fact, results
ambucil) in combination with corticosteroids have been shown to of the study can be explained by a lag in anti-PLA2R antibody decline
be effective in the treatment of patients with MN (150,151). The in the tacrolimus during the first 6 months versus a faster decrease in
significant toxicities associated with alkylating agents constitute a anti-PLA2R antibody decline in the cyclophosphamide arm.
major concern. However, in patients presenting with an already low Another trial was the RI-CYCLO trial in which 74 patients were
GFR or deteriorating kidney function, a course of cyclophospha- randomized to receive 6 months of cyclophosphamide alternating
mide combined with prednisone may be considered. Alternatively, with corticosteroids versus two doses of rituximab (1 g each) given on
CNIs have been shown to be effective in treating patients with MN. day 1 and day 15. The primary outcome was probability of complete
Both tacrolimus and cyclosporine have been used either as mono- or partial remission at 12 months. There were few partial and com-
therapy or in combination with corticosteroids, with good results plete remissions at 12 months in patients receiving rituximab com-
(152–156). The major limitation with the use of CNIs is the high pared with cyclophosphamide, but this was not statistically significant.
rate of relapse after discontinuation of therapy (155,157), which At 24 months there were similar rates of complete and partial remis-
results in long-term use of CNIs in such patients and can in turn sions in the rituximab (85%) and cyclophosphamide (81%) groups.
lead to nephrotoxicity. This rate of complete remission and partial remission remained similar
Over the past decade, rituximab has emerged as the most prom- at 36 months. There were similar rates of adverse events in both
ising therapy in patients with MN. Since the initial report of suc- groups during the study period with infusion-related reactions more
cessful treatment of MN patients with rituximab, several additional common in the rituximab group and leukopenia and infectious com-
prospective trials have emerged and have confirmed the effectiveness plications more common in the cyclophosphamide group. Based on
of rituximab (149,158–160). This was ultimately followed by Mem- this trial, rituximab and cyclophosphamide combined with corticoster-
branous Nephropathy Trial of Rituximab (MENTOR), a random- oids had a similar safety and efficacy profile.
ized, open-label, noninferiority trial comparing rituximab with
cyclosporine therapy. This study enrolled 130 patients with MN, at
moderate risk of progression, with at least 5 g/d of proteinuria and 4 Although the STARMEN trial showed increased
eGFR .40 ml/min per 1.73 m2. Patients were randomized to receive remission with methylprednisolone/cyclophospha-
either rituximab 1 g for two doses (a second identical course of rituxi-
mab was given to patients that had .25% decrease in proteinuria at
mide compared with tacrolimus and low-dose
6 months) or cyclosporine 3.5 mg/kg for 12 months. Rituximab was rituximab, the late administration and low dose of
noninferior to cyclosporine in inducing remission at 12 months but rituximab render the trial an inconclusive compari-
was superior to cyclosporine in maintaining proteinuria remission at son between cyclophosphamide and rituximab.
24 months. The rates of complete and partial remission at 24 months
differed widely, with remission in 60% of rituximab-treated versus 4 The RI-CYCLO trial showed comparable remission
20% of cyclosporine-treated patients (161). Among patients who expe- at 24 and 36 months in patients treated with
rienced complete or partial remission, those who received cyclosporine cyclophosphamide versus rituximab.
had lower creatinine clearance at 24 months (87 ml/min per 1.73 m2)
compared with those who received rituximab (100 ml/min per
1.73 m2), despite having not taken cyclosporine for 12 months Finally, in patients who become sensitized to rituximab (a chime-
(161). This highlights the fact that the effect of CNIs on renal func- ric type I anti-CD20 agent), a fully humanized type I anti-CD20
tion can be long lasting even after discontinuation of therapy. Given drug such as ofatumumab has been used successfully (164). Despite
these results, the upcoming Kidney Disease: Improving Global Out- the effectiveness of rituximab in inducing remission in patients with
comes guidelines recommend rituximab as first-line therapy (60). MN, up to 30% of patients do not achieve remission. A case series
Whether rituximab or cyclophosphamide is more efficacious in from Mayo Clinic reported on three patients with PLA2R-positive
treating patients with MN has been a subject of ongoing debate in MN who did not respond to rituximab. Obinutuzumab is a type II
the nephrology community. There are currently two trials that have anti-CD20 agent, which leads to more potent B cell depletion and
compared cyclophosphamide in combination with prednisone to may have a greater effect on memory B cells than rituximab. After
rituximab (162,163). First was the STARMEN trial in which treatment with obinutuzumab, two patients experienced complete
86 patients were randomized to receive either methylprednisolone remission and one patient partial remission. This suggests that obinu-
on months 1, 3, and 5 and cyclophosphamide on months 2, 4, and tuzumab may be an alternative for patients who do not experience
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 347

clinical or immunologic remission with rituximab (93,165). There is randomized trial indicates initial prednisolone treatment for childhood
currently an ongoing trial comparing the safety and efficacy of obinu- nephrotic syndrome for two months is not inferior to six-month treat-
ment. Kidney Int 87: 225–232, 2015 PubMed
tuzumab with tacrolimus therapy (NCT04629248).
20. Sinha A, Saha A, Kumar M, Sharma S, Afzal K, Mehta A, et al.:
Extending initial prednisolone treatment in a randomized control trial
References from 3 to 6 months did not significantly influence the course of illness
1. Glassock RJ, Fervenza FC, Hebert L, Cameron JS: Nephrotic syndrome in children with steroid-sensitive nephrotic syndrome. Kidney Int 87:
redux. Nephrol Dial Transplant 30: 12–17, 2015 PubMed 217–224, 2015 PubMed
2. van de Logt AE, Rijpma SR, Vink CH, Prudon-Rosmulder E, Wetzels 21. Webb NJA, Woolley RL, Lambe T, Frew E, Brettell EA, Barsoum EN,
JF, van Berkel M: The bias between different albumin assays may affect et al.; PREDNOS Collaborative Group: Long term tapering versus stan-
clinical decision-making. Kidney Int 95: 1514–1517, 2019 PubMed dard prednisolone treatment for first episode of childhood nephrotic
3. Meyrier A, Niaudet P: Acute kidney injury complicating nephrotic syn- syndrome: Phase III randomised controlled trial and economic evalua-
drome of minimal change disease. Kidney Int 94: 861–869, 2018 tion. BMJ 365: l1800, 2019 PubMed
PubMed 22. Hahn D, Hodson EM, Willis NS, Craig JC: Corticosteroid therapy for
4. Glassock RJ: Secondary minimal change disease. Nephrol Dial Trans- nephrotic syndrome in children. Cochrane Database Syst Rev (3):
plant 18[Suppl 6]: vi52–vi58, 2003 PubMed CD001533, 2015 PubMed
5. Shalhoub RJ: Pathogenesis of lipoid nephrosis: A disorder of T-cell 23. Waldman M, Crew RJ, Valeri A, Busch J, Stokes B, Markowitz G,
function. Lancet 2: 556–560, 1974 PubMed et al.: Adult minimal-change disease: Clinical characteristics, treatment,
6. Lagrue G, Xheneumont S, Branellec A, Hirbec G, Weil B: A vascular per- and outcomes. Clin J Am Soc Nephrol 2: 445–453, 2007 PubMed
meability factor elaborated from lymphocytes. I. Demonstration in patients 24. Li H, Shi X, Shen H, Li X, Wang H, Li H, et al.: Tacrolimus versus
with nephrotic syndrome. Biomedicine (Paris) 23: 37–40, 1975 PubMed intravenous pulse cyclophosphamide therapy in Chinese adults with
7. Heslan JM, Branellec AI, Pilatte Y, Lang P, Lagrue G: Differentiation steroid-resistant idiopathic minimal change nephropathy: A multicenter,
between vascular permeability factor and IL-2 in lymphocyte superna- open-label, nonrandomized cohort trial. Clin Ther 34: 1112–1120,
tants from patients with minimal-change nephrotic syndrome. Clin Exp 2012 PubMed
Immunol 86: 157–162, 1991 PubMed 25. Ponticelli C, Edefonti A, Ghio L, Rizzoni G, Rinaldi S, Gusmano R,
8. Cheung PK, Stulp B, Immenschuh S, Borghuis T, Baller JF, Bakker et al.: Cyclosporin versus cyclophosphamide for patients with steroid-
WW: Is 100KF an isoform of hemopexin? Immunochemical characteri- dependent and frequently relapsing idiopathic nephrotic syndrome: A
zation of the vasoactive plasma factor 100KF. J Am Soc Nephrol 10: multicentre randomized controlled trial. Nephrol Dial Transplant 8:
1700–1708, 1999 PubMed 1326–1332, 1993 PubMed
9. Cheung PK, Klok PA, Baller JF, Bakker WW: Induction of experimen- 26. Li X, Li H, Chen J, He Q, Lv R, Lin W, et al.: Tacrolimus as a
tal proteinuria in vivo following infusion of human plasma hemopexin. steroid-sparing agent for adults with steroid-dependent minimal change
Kidney Int 57: 1512–1520, 2000 PubMed nephrotic syndrome. Nephrol Dial Transplant 23: 1919–1925, 2008
10. Clement LC, Avila-Casado C, Mace C, Soria E, Bakker WW, Kersten PubMed
S, et al.: Podocyte-secreted angiopoietin-like-4 mediates proteinuria in 27. Meyrier A, Condamin MC, Broneer D; Collaborative Group of the French
glucocorticoid-sensitive nephrotic syndrome. Nat Med 17: 117–122, Society of Nephrology: Treatment of adult idiopathic nephrotic syndrome
2011 PubMed with cyclosporin A: Minimal-change disease and focal-segmental glomerulo-
11. Lai KW, Wei CL, Tan LK, Tan PH, Chiang GS, Lee CG, et al.: Over- sclerosis. Clin Nephrol 35[Suppl 1]: S37–S42, 1991 PubMed
expression of interleukin-13 induces minimal-change-like nephropathy 28. Li X, Liu Z, Wang L, Wang R, Ding G, Shi W, et al.: Tacrolimus
in rats. J Am Soc Nephrol 18: 1476–1485, 2007 PubMed monotherapy after intravenous methylprednisolone in adults with mini-
12. Chugh SS, Clement LC, Mace C: New insights into human minimal mal change nephrotic syndrome. J Am Soc Nephrol 28: 1286–1295,
change disease: Lessons from animal models. Am J Kidney Dis 59: 284– 2017 PubMed
292, 2012 PubMed 29. Medjeral-Thomas NR, Lawrence C, Condon M, Sood B, Warwicker P,
13. Garin EH, Blanchard DK, Matsushima K, Djeu JY: IL-8 production by Brown H, et al.: Randomized, controlled trial of tacrolimus and pred-
peripheral blood mononuclear cells in nephrotic patients. Kidney Int 45: nisolone monotherapy for adults with de novo minimal change disease:
1311–1317, 1994 PubMed A multicenter, randomized, controlled trial. Clin J Am Soc Nephrol 15:
14. Kondo S, Yoshizawa N, Kusumi Y, Takeuchi A, Torikata C: Studies of 209–218, 2020 PubMed
glomerular permeability factor (GPF) in focal segmental glomerular 30. Gellermann J, Weber L, Pape L, T€onshoff B, Hoyer P, Querfeld U;
sclerosis and the relationship between GPF and vascular permeability Gesellschaft f€ ur P€adiatrische Nephrologie (GPN): Mycophenolate mofe-
factor (VPF). Clin Nephrol 52: 278–284, 1999 PubMed til versus cyclosporin A in children with frequently relapsing nephrotic
15. Clement LC, Mace C, Avila-Casado C, Joles JA, Kersten S, Chugh SS: syndrome. J Am Soc Nephrol 24: 1689–1697, 2013 PubMed
Circulating angiopoietin-like 4 links proteinuria with hypertriglyceride- 31. Geng HY, Ji LN, Chen CY, Tu J, Li HR, Bao R, et al.: [Mycophenolate
mia in nephrotic syndrome. Nat Med 20: 37–46, 2014 PubMed mofetil versus cyclosporine A in children with primary refractory nephrotic
16. Kim AH, Chung JJ, Akilesh S, Koziell A, Jain S, Hodgin JB, et al.: B syndrome]. Zhonghua Er Ke Za Zhi 56: 651–656, 2018 PubMed
cell-derived IL-4 acts on podocytes to induce proteinuria and foot pro- 32. Sinha A, Puraswani M, Kalaivani M, Goyal P, Hari P, Bagga A: Effi-
cess effacement. JCI Insight 2: e81836, 2017 PubMed cacy and safety of mycophenolate mofetil versus levamisole in frequently
17. Boumediene A, Vachin P, Sendeyo K, Oniszczuk J, Zhang SY, Henique relapsing nephrotic syndrome: An open-label randomized controlled
C, et al.: NEPHRUTIX: A randomized, double-blind, placebo vs trial. Kidney Int 95: 210–218, 2019 PubMed
rituximab-controlled trial assessing T-cell subset changes in minimal 33. Remy P, Audard V, Natella PA, Pelle G, Dussol B, Leray-Moragues H,
change nephrotic syndrome. J Autoimmun 88: 91–102, 2018 PubMed et al.; MSN Trial Investigators: An open-label randomized controlled trial
18. Watts AJB, Keller KH, Lerner G, Rosales I, Collins AB, Sekulic M, of low-dose corticosteroid plus enteric-coated mycophenolate sodium ver-
et al.: Discovery of autoantibodies targeting nephrin in minimal change sus standard corticosteroid treatment for minimal change nephrotic syn-
disease supports a novel autoimmune etiology. J Am Soc Nephrol 33: drome in adults (MSN study). Kidney Int 94: 1217–1226, 2018 PubMed
238–252, 2022 PubMed 34. Benz K, D€otsch J, Rascher W, Stachel D: Change of the course of
19. Yoshikawa N, Nakanishi K, Sako M, Oba MS, Mori R, Ota E, et al.; steroid-dependent nephrotic syndrome after rituximab therapy. Pediatr
Japanese Study Group of Kidney Disease in Children: A multicenter Nephrol 19: 794–797, 2004 PubMed
348 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

35. Ravani P, Rossi R, Bonanni A, Quinn RR, Sica F, Bodria M, et al.: 51. Wang CS, Travers C, McCracken C, Leong T, Gbadegesin R, Quiroga
Rituximab in children with steroid-dependent nephrotic syndrome: A, et al.: Adrenocorticotropic hormone for childhood nephrotic syn-
A multicenter, open-label, noninferiority, randomized controlled trial. drome: The ATLANTIS randomized trial. Clin J Am Soc Nephrol 13:
J Am Soc Nephrol 26: 2259–2266, 2015 PubMed 1859–1865, 2018 PubMed
36. Iijima K, Sako M, Nozu K, Mori R, Tuchida N, Kamei K, et al.; Ritux- 52. Szeto CC, Lai FM, Chow KM, Kwan BC, Kwong VW, Leung CB,
imab for Childhood-onset Refractory Nephrotic Syndrome (RCRNS) et al.: Long-term outcome of biopsy-proven minimal change nephropa-
Study Group: Rituximab for childhood-onset, complicated, frequently thy in Chinese adults. Am J Kidney Dis 65: 710–718, 2015 PubMed
relapsing nephrotic syndrome or steroid-dependent nephrotic syndrome: 53. Sethi S, Glassock RJ, Fervenza FC: Focal segmental glomerulosclerosis:
A multicentre, double-blind, randomised, placebo-controlled trial. Lan- Towards a better understanding for the practicing nephrologist. Nephrol
cet 384: 1273–1281, 2014 PubMed Dial Transplant 30: 375–384, 2015 PubMed
37. Ruggenenti P, Ruggiero B, Cravedi P, Vivarelli M, Massella L, Marasa 54. Izzedine H, Launay-Vacher V, Bourry E, Brocheriou I, Karie S, Deray
M, et al.; Rituximab in Nephrotic Syndrome of Steroid-Dependent or G: Drug-induced glomerulopathies. Expert Opin Drug Saf 5: 95–106,
Frequently Relapsing Minimal Change Disease Or Focal Segmental 2006 PubMed
Glomerulosclerosis (NEMO) Study Group: Rituximab in steroid- 55. Chandra P, Kopp JB: Viruses and collapsing glomerulopathy: A brief
dependent or frequently relapsing idiopathic nephrotic syndrome. J Am critical review. Clin Kidney J 6: 1–5, 2013 PubMed
Soc Nephrol 25: 850–863, 2014 PubMed 56. Kambham N, Markowitz GS, Valeri AM, Lin J, D’Agati VD: Obesity-
38. Webb H, Jaureguiberry G, Dufek S, Tullus K, Bockenhauer D: Cyclo- related glomerulopathy: An emerging epidemic. Kidney Int 59: 1498–
phosphamide and rituximab in frequently relapsing/steroid-dependent 1509, 2001 PubMed
nephrotic syndrome. Pediatr Nephrol 31: 589–594, 2016 PubMed 57. Kriz W, Lemley KV: A potential role for mechanical forces in the
39. Basu B, Sander A, Roy B, Preussler S, Barua S, Mahapatra TKS, et al.: detachment of podocytes and the progression of CKD. J Am Soc
Efficacy of rituximab vs tacrolimus in pediatric corticosteroid-dependent Nephrol 26: 258–269, 2015 PubMed
nephrotic syndrome: A randomized clinical trial. JAMA Pediatr 172: 58. Kriz W, Lemley KV: Potential relevance of shear stress for slit diaphragm
757–764, 2018 PubMed and podocyte function. Kidney Int 91: 1283–1286, 2017 PubMed
40. Hogan J, Dossier C, Kwon T, Macher MA, Maisin A, Couderc A, et al.: 59. De Vriese AS, Wetzels JF, Glassock RJ, Sethi S, Fervenza FC: Thera-
Effect of different rituximab regimens on B cell depletion and time to peutic trials in adult FSGS: Lessons learned and the road forward. Nat
relapse in children with steroid-dependent nephrotic syndrome. Pediatr Rev Nephrol 17: 619–630, 2021 PubMed
Nephrol 34: 253–259, 2019 PubMed 60. Rovin BH, Adler SG, Barratt J, Bridoux F, Burdge KA, Chan TM,
41. Chan EY, Webb H, Yu E, Ghiggeri GM, Kemper MJ, Ma AL, et al.: et al.: Executive summary of the KDIGO 2021 guideline for the man-
Both the rituximab dose and maintenance immunosuppression in ste- agement of glomerular diseases. Kidney Int 100: 753–779, 2021
roid-dependent/frequently-relapsing nephrotic syndrome have important PubMed
effects on outcomes. Kidney Int 97: 393–401, 2020 PubMed 61. Campbell KN, Tumlin JA: Protecting podocytes: A key target for ther-
42. Ravani P, Magnasco A, Edefonti A, Murer L, Rossi R, Ghio L, et al.: apy of focal segmental glomerulosclerosis. Am J Nephrol 47[Suppl 1]:
Short-term effects of rituximab in children with steroid- and 14–29, 2018 PubMed
calcineurin-dependent nephrotic syndrome: A randomized controlled 62. Campbell KN, Wong JS, Gupta R, Asanuma K, Sudol M, He JC, et al.:
trial. Clin J Am Soc Nephrol 6: 1308–1315, 2011 PubMed Yes-associated protein (YAP) promotes cell survival by inhibiting proapopto-
43. Ravani P, Ponticelli A, Siciliano C, Fornoni A, Magnasco A, Sica F, tic dendrin signaling. J Biol Chem 288: 17057–17062, 2013 PubMed
et al.: Rituximab is a safe and effective long-term treatment for children 63. Schwartzman M, Reginensi A, Wong JS, Basgen JM, Meliambro K,
with steroid and calcineurin inhibitor-dependent idiopathic nephrotic Nicholas SB, et al.: Podocyte-specific deletion of Yes-associated protein
syndrome. Kidney Int 84: 1025–1033, 2013 PubMed causes FSGS and progressive renal failure. J Am Soc Nephrol 27: 216–
44. Magnasco A, Ravani P, Edefonti A, Murer L, Ghio L, Belingheri M, 226, 2016 PubMed
et al.: Rituximab in children with resistant idiopathic nephrotic syn- 64. Wharram BL, Goyal M, Wiggins JE, Sanden SK, Hussain S, Filipiak
drome. J Am Soc Nephrol 23: 1117–1124, 2012 PubMed WE, et al.: Podocyte depletion causes glomerulosclerosis: Diphtheria
45. Ravani P, Bonanni A, Ghiggeri GM: Randomised controlled trial com- toxin-induced podocyte depletion in rats expressing human diphtheria
paring ofatumumab to rituximab in children with steroid-dependent toxin receptor transgene. J Am Soc Nephrol 16: 2941–2952, 2005
and calcineurin inhibitor-dependent idiopathic nephrotic syndrome: PubMed
Study protocol. BMJ Open 7: e013319, 2017 PubMed 65. Eng DG, Sunseri MW, Kaverina NV, Roeder SS, Pippin JW, Shank-
46. Iwabuchi Y, Miyabe Y, Makabe S, Nakano M, Manabe S, Karasawa K, land SJ: Glomerular parietal epithelial cells contribute to adult podocyte
et al.: Comparison of the response of frequently relapsing steroid- regeneration in experimental focal segmental glomerulosclerosis. Kidney
dependent minimal change nephrotic syndrome to rituximab therapy Int 88: 999–1012, 2015 PubMed
between childhood-onset and adult-onset disease. Medicine (Baltimore) 66. Kretzler M: Role of podocytes in focal sclerosis: Defining the point of
97: e12704, 2018 PubMed no return. J Am Soc Nephrol 16: 2830–2832, 2005 PubMed
47. Fenoglio R, Sciascia S, Beltrame G, Mesiano P, Ferro M, Quattrocchio 67. Savin VJ, Sharma M, Zhou J, Gennochi D, Fields T, Sharma R, et al.:
G, et al.: Rituximab as a front-line therapy for adult-onset minimal Renal and hematological effects of CLCF-1, a B-cell-stimulating cyto-
change disease with nephrotic syndrome. Oncotarget 9: 28799–28804, kine of the IL-6 family. J Immunol Res 2015: 714964, 2015 PubMed
2018 PubMed 68. Wei C, El Hindi S, Li J, Fornoni A, Goes N, Sageshima J, et al.: Circu-
48. Kronbichler A, Kerschbaum J, Fernandez-Fresnedo G, Hoxha E, lating urokinase receptor as a cause of focal segmental glomerulosclero-
Kurschat CE, Busch M, et al.: Rituximab treatment for relapsing mini- sis. Nat Med 17: 952–960, 2011 PubMed
mal change disease and focal segmental glomerulosclerosis: A systematic 69. Delville M, Sigdel TK, Wei C, Li J, Hsieh SC, Fornoni A, et al.: A circu-
review. Am J Nephrol 39: 322–330, 2014 PubMed lating antibody panel for pretransplant prediction of FSGS recurrence
49. Kittanamongkolchai W, Cheungpasitporn W, Zand L: Efficacy and after kidney transplantation. Sci Transl Med 6: 256ra136, 2014 PubMed
safety of adrenocorticotropic hormone treatment in glomerular diseases: 70. Wada T, Nangaku M: A circulating permeability factor in focal
A systematic review and meta-analysis. Clin Kidney J 9: 387–396, 2016 segmental glomerulosclerosis: The hunt continues. Clin Kidney J 8:
PubMed 708–715, 2015 PubMed
50. Madan A, Mijovic-Das S, Stankovic A, Teehan G, Milward AS, Khast- 71. Lepori N, Zand L, Sethi S, Fernandez-Juarez G, Fervenza FC: Clinical
gir A: Acthar gel in the treatment of nephrotic syndrome: A multicenter and pathological phenotype of genetic causes of focal segmental glomer-
retrospective case series. BMC Nephrol 17: 37, 2016 PubMed ulosclerosis in adults. Clin Kidney J 11: 179–190, 2018 PubMed
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 349

72. De Vriese AS, Sethi S, Nath KA, Glassock RJ, Fervenza FC: Differenti- 90. Kronbichler A, Gauckler P, Bruchfeld A: Rituximab in minimal change
ating primary, genetic, and secondary FSGS in adults: A clinicopatho- disease and focal segmental glomerulosclerosis. Nephrol Dial Transplant
logic approach. J Am Soc Nephrol 29: 759–774, 2018 PubMed 36: 983–985, 2021 PubMed
73. Buxeda A, Said S, Nasr SH, Soler MJ, Howard MT, Maguire LJ, et al.: 91. Roccatello D, Sciascia S, Rossi D, Alpa M, Naretto C, Radin M, et al.:
Crystal-induced podocytopathy producing collapsing focal segmental High-dose rituximab ineffective for focal segmental glomerulosclerosis: A
glomerulosclerosis in monoclonal gammopathy of renal significance: long-term observation study. Am J Nephrol 46: 108–113, 2017 PubMed
A case report. Kidney Med 3: 659–664, 2021 PubMed 92. Furie RA, Aroca G, Cascino MD, Garg JP, Rovin BH, Alvarez A, et al.:
74. Sethi S, Zand L, Nasr SH, Glassock RJ, Fervenza FC: Focal and B-cell depletion with obinutuzumab for the treatment of proliferative
segmental glomerulosclerosis: Clinical and kidney biopsy correlations. lupus nephritis: A randomised, double-blind, placebo-controlled trial.
Clin Kidney J 7: 531–537, 2014 PubMed Ann Rheum Dis 81: 100–107, 2022 PubMed
75. Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Chary- 93. Klomjit N, Fervenza FC, Zand L: Successful treatment of patients with
tan DM, et al.; CREDENCE Trial Investigators: Canagliflozin and refractory PLA2R-associated membranous nephropathy with obinutuzu-
renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 380: mab: A report of 3 cases. Am J Kidney Dis 76: 883–888, 2020 PubMed
2295–2306, 2019 PubMed 94. Gipson DS, Trachtman H, Kaskel FJ, Greene TH, Radeva MK, Gass-
76. van den Berg JG, van den Bergh Weerman MA, Assmann KJ, Weening man JJ, et al.: Clinical trial of focal segmental glomerulosclerosis in chil-
JJ, Florquin S: Podocyte foot process effacement is not correlated with dren and young adults. Kidney Int 80: 868–878, 2011 PubMed
the level of proteinuria in human glomerulopathies. Kidney Int 66: 95. Hogan J, Bomback AS, Mehta K, Canetta PA, Rao MK, Appel GB,
1901–1906, 2004 PubMed et al.: Treatment of idiopathic FSGS with adrenocorticotropic hormone
77. Deegens JK, Dijkman HB, Borm GF, Steenbergen EJ, van den Berg gel. Clin J Am Soc Nephrol 8: 2072–2081, 2013 PubMed
JG, Weening JJ, et al.: Podocyte foot process effacement as a diagnostic 96. Bomback AS, Canetta PA, Beck LH Jr, Ayalon R, Radhakrishnan J, Appel
tool in focal segmental glomerulosclerosis. Kidney Int 74: 1568–1576, GB: Treatment of resistant glomerular diseases with adrenocorticotropic
2008 PubMed hormone gel: A prospective trial. Am J Nephrol 36: 58–67, 2012 PubMed
78. Maas RJ, Deegens JK, Smeets B, Moeller MJ, Wetzels JF: Minimal 97. Trachtman H, Nelson P, Adler S, Campbell KN, Chaudhuri A, Dere-
change disease and idiopathic FSGS: Manifestations of the same disease. bail VK, et al.; DUET Study Group: DUET: A phase 2 study evaluat-
Nat Rev Nephrol 12: 768–776, 2016 PubMed ing the efficacy and safety of sparsentan in patients with FSGS. J Am
79. Hommos MS, De Vriese AS, Alexander MP, Sethi S, Vaughan L, Zand L, Soc Nephrol 29: 2745–2754, 2018 PubMed
et al.: The incidence of primary vs secondary focal segmental glomerulosclero- 98. Heerspink HJL, Stefansson BV, Correa-Rotter R, Chertow GM, Greene
sis: A clinicopathologic study. Mayo Clin Proc 92: 1772–1781, 2017 PubMed T, Hou FF, et al.; DAPA-CKD Trial Committees and Investigators:
80. Zand L, Glassock RJ, De Vriese AS, Sethi S, Fervenza FC: What are we Dapagliflozin in patients with chronic kidney disease. N Engl J Med
missing in the clinical trials of focal segmental glomerulosclerosis? 383: 1436–1446, 2020 PubMed
Nephrol Dial Transplant 32[Suppl 1]: i14–i21, 2017 PubMed 99. Wheeler DC, Jongs N, Stefansson BV, Chertow GM, Greene T, Hou
81. Hogan J, Mohan P, Appel GB: Diagnostic tests and treatment options FF, et al.; DAPA-CKD Trial Committees and Investigators: Safety and
in glomerular disease: 2014 update. Am J Kidney Dis 63: 656–666, efficacy of dapagliflozin in patients with focal segmental glomerulo-
2014 PubMed sclerosis: A prespecified analysis of the dapagliflozin and prevention of
82. Rood IM, Bavinck A, Lipska-ZieR tkiewicz BS, Lugtenberg D, Schaefer F, adverse outcomes in chronic kidney disease (DAPA-CKD) trial. Nephrol
Deegens JKJ, et al.: Later response to corticosteroids in adults with pri- Dial Transplant 37: 1647–1656, 2022 PubMed
mary focal segmental glomerular sclerosis is associated with favorable 100. Santın S, Bullich G, Tazon-Vega B, Garcıa-Maset R, Gimenez I, Silva I,
outcomes. Kidney Int Rep 7: 87–98, 2021 PubMed et al.: Clinical utility of genetic testing in children and adults with
83. Cattran DC, Appel GB, Hebert LA, Hunsicker LG, Pohl MA, Hoy steroid-resistant nephrotic syndrome. Clin J Am Soc Nephrol 6: 1139–
WE, et al.; North America Nephrotic Syndrome Study Group: A ran- 1148, 2011 PubMed
domized trial of cyclosporine in patients with steroid-resistant focal seg- 101. Gribouval O, Boyer O, Hummel A, Dantal J, Martinez F, Sberro-Sous-
mental glomerulosclerosis. Kidney Int 56: 2220–2226, 1999 PubMed san R, et al.: Identification of genetic causes for sporadic steroid-
84. Ramachandran R, Kumar V, Rathi M, Nada R, Jha V, Gupta KL, et al.: resistant nephrotic syndrome in adults. Kidney Int 94: 1013–1022,
Tacrolimus therapy in adult-onset steroid-resistant nephrotic syndrome 2018 PubMed
due to a focal segmental glomerulosclerosis single-center experience. 102. Landini S, Mazzinghi B, Becherucci F, Allinovi M, Provenzano A,
Nephrol Dial Transplant 29: 1918–1924, 2014 PubMed Palazzo V, et al.: Reverse phenotyping after whole-exome sequencing in
85. Laurin LP, Gasim AM, Poulton CJ, Hogan SL, Jennette JC, Falk RJ, steroid-resistant nephrotic syndrome. Clin J Am Soc Nephrol 15: 89–100,
et al.: Treatment with glucocorticoids or calcineurin inhibitors in pri- 2020 PubMed
mary FSGS. Clin J Am Soc Nephrol 11: 386–394, 2016 PubMed 103. Warejko JK, Tan W, Daga A, Schapiro D, Lawson JA, Shril S, et al.:
86. Faul C, Donnelly M, Merscher-Gomez S, Chang YH, Franz S, Whole exome sequencing of patients with steroid-resistant nephrotic
Delfgaauw J, et al.: The actin cytoskeleton of kidney podocytes is a syndrome. Clin J Am Soc Nephrol 13: 53–62, 2018 PubMed
direct target of the antiproteinuric effect of cyclosporine A. Nat Med 104. Miao J, Pinto E Vairo F, Hogan MC, Erickson SB, El Ters M, Bentall
14: 931–938, 2008 PubMed AJ, et al.: Identification of genetic causes of focal segmental glomerulo-
87. Meyrier A, No€el LH, Auriche P, Callard P; Collaborative Group of the sclerosis increases with proper patient selection. Mayo Clin Proc 96:
Societe de Nephrologie: Long-term renal tolerance of cyclosporin A 2342–2353, 2021 PubMed
treatment in adult idiopathic nephrotic syndrome. Kidney Int 45: 105. Groopman EE, Marasa M, Cameron-Christie S, Petrovski S, Aggarwal VS,
1446–1456, 1994 PubMed Milo-Rasouly H, et al.: Diagnostic utility of exome sequencing for kidney
88. DaSilva I, Huerta A, Quintana L, Redondo B, Iglesias E, Draibe J, disease. N Engl J Med 380: 142–151, 2019 PubMed
et al.; Spanish Group for the Study of Glomerular Diseases (GLOSEN): 106. Malone AF, Phelan PJ, Hall G, Cetincelik U, Homstad A, Alonso AS,
Rituximab for steroid-dependent or frequently relapsing idiopathic et al.: Rare hereditary COL4A3/COL4A4 variants may be mistaken for
nephrotic syndrome in adults: A retrospective, multicenter study in familial focal segmental glomerulosclerosis. Kidney Int 86: 1253–1259,
Spain. BioDrugs 31: 239–249, 2017 PubMed 2014 PubMed
89. Tedesco M, Mescia F, Pisani I, Allinovi M, Casazza G, Del Vecchio L, 107. Gast C, Pengelly RJ, Lyon M, Bunyan DJ, Seaby EG, Graham N, et al.:
et al.; Italian Society of Nephrology Immunopathology Working Collagen (COL4A) mutations are the most frequent mutations underly-
Group: The role of rituximab in primary focal segmental glomerular ing adult focal segmental glomerulosclerosis. Nephrol Dial Transplant
sclerosis of the adult. Kidney Int Rep 7: 1878–1886, 2022 PubMed 31: 961–970, 2016 PubMed
350 Nephrology Self-Assessment Program - Vol 21, No 5, December 2022

108. Sen ES, Dean P, Yarram-Smith L, Bierzynska A, Woodward G, Buxton C, 126. Larsen CP, Messias NC, Silva FG, Messias E, Walker PD: Determina-
et al.: Clinical genetic testing using a custom-designed steroid-resistant tion of primary versus secondary membranous glomerulopathy utilizing
nephrotic syndrome gene panel: Analysis and recommendations. J Med phospholipase A2 receptor staining in renal biopsies. Mod Pathol 26:
Genet 54: 795–804, 2017 PubMed 709–715, 2013 PubMed
109. Yao T, Udwan K, John R, Rana A, Haghighi A, Xu L, et al.: Integra- 127. Stehle T, Audard V, Ronco P, Debiec H: Phospholipase A2 receptor
tion of genetic testing and pathology for the diagnosis of adults with and sarcoidosis-associated membranous nephropathy. Nephrol Dial
FSGS. Clin J Am Soc Nephrol 14: 213–223, 2019 PubMed Transplant 30: 1047–1050, 2015 PubMed
110. B€uscher AK, Beck BB, Melk A, Hoefele J, Kranz B, Bamborschke D, 128. Xie Q, Li Y, Xue J, Xiong Z, Wang L, Sun Z, et al.: Renal phospholi-
et al.; German Pediatric Nephrology Association (GPN): Rapid pase A2 receptor in hepatitis B virus-associated membranous nephropa-
response to cyclosporin A and favorable renal outcome in nongenetic thy. Am J Nephrol 41: 345–353, 2015 PubMed
versus genetic steroid-resistant nephrotic syndrome. Clin J Am Soc 129. Behnert A, Schiffer M, M€ uller-Deile J, Beck LH Jr, Mahler M, Fritzler
Nephrol 11: 245–253, 2016 PubMed MJ: Antiphospholipase A2 receptor autoantibodies: A comparison of
111. Hoxha E, Reinhard L, Stahl RAK: Membranous nephropathy: New three different immunoassays for the diagnosis of idiopathic membra-
pathogenic mechanisms and their clinical implications. Nat Rev Nephrol nous nephropathy. J Immunol Res 2014: 143274, 2014 PubMed
18: 466–478, 2022 PubMed 130. Timmermans SA, Abdul Hamid MA, Cohen Tervaert JW, Damoiseaux
112. Bobart SA, Tehranian S, Sethi S, Alexander MP, Nasr SH, Moura JG, van Paassen P; Limburg Renal Registry: Anti-PLA2R antibodies
Marta C, et al.: A target antigen-based approach to the classification as a prognostic factor in PLA2R-related membranous nephropathy.
of membranous nephropathy. Mayo Clin Proc 96: 577–591, 2021 Am J Nephrol 42: 70–77, 2015 PubMed
PubMed 131. Hoxha E, Harendza S, Pinnschmidt H, Panzer U, Stahl RA: PLA2R anti-
113. Beck LH Jr, Bonegio RG, Lambeau G, Beck DM, Powell DW, Cum- body levels and clinical outcome in patients with membranous nephropa-
mins TD, et al.: M-type phospholipase A2 receptor as target antigen in thy and non-nephrotic range proteinuria under treatment with inhibitors
idiopathic membranous nephropathy. N Engl J Med 361: 11–21, 2009 of the renin-angiotensin system. PLoS One 9: e110681, 2014 PubMed
PubMed 132. Kanigicherla D, Gummadova J, McKenzie EA, Roberts SA, Harris S,
114. Tomas NM, Beck LH Jr, Meyer-Schwesinger C, Seitz-Polski B, Ma H, Nikam M, et al.: Anti-PLA2R antibodies measured by ELISA predict
Zahner G, et al.: Thrombospondin type-1 domain-containing 7A in idi- long-term outcome in a prevalent population of patients with idiopathic
opathic membranous nephropathy. N Engl J Med 371: 2277–2287, membranous nephropathy. Kidney Int 83: 940–948, 2013 PubMed
2014 PubMed 133. Hoxha E, Harendza S, Pinnschmidt H, Panzer U, Stahl RA: M-type
115. Sethi S, Debiec H, Madden B, Charlesworth MC, Morelle J, Gross L, phospholipase A2 receptor autoantibodies and renal function in patients
et al.: Neural epidermal growth factor-like 1 protein (NELL-1) associ- with primary membranous nephropathy. Clin J Am Soc Nephrol 9:
ated membranous nephropathy. Kidney Int 97: 163–174, 2020 PubMed 1883–1890, 2014 PubMed
116. Sethi S, Debiec H, Madden B, Vivarelli M, Charlesworth MC, Ravin- 134. Burbelo PD, Joshi M, Chaturvedi A, Little DJ, Thurlow JS, Waldman
dran A, et al.: Semaphorin 3B-associated membranous nephropathy is a M, et al.: Detection of PLA2R autoantibodies before the diagnosis of
distinct type of disease predominantly present in pediatric patients. Kid- membranous nephropathy. J Am Soc Nephrol 31: 208–217, 2020
ney Int 98: 1253–1264, 2020 PubMed PubMed
117. Sethi S, Madden B, Debiec H, Morelle J, Charlesworth MC, Gross L, 135. Hoxha E, Thiele I, Zahner G, Panzer U, Harendza S, Stahl RA:
et al.: Protocadherin 7-associated membranous nephropathy. J Am Soc Phospholipase A2 receptor autoantibodies and clinical outcome in
Nephrol 32: 1249–1261, 2021 PubMed patients with primary membranous nephropathy. J Am Soc Nephrol 25:
118. Al-Rabadi LF, Caza T, Trivin-Avillach C, Rodan AR, Andeen N, 1357–1366, 2014 PubMed
Hayashi N, et al.: Serine protease HTRA1 as a novel target antigen in 136. Radice A, Trezzi B, Maggiore U, Pregnolato F, Stellato T, Napodano
primary membranous nephropathy. J Am Soc Nephrol 32: 1666–1681, P, et al.: Clinical usefulness of autoantibodies to M-type phospholipase
2021 PubMed A2 receptor (PLA2R) for monitoring disease activity in idiopathic mem-
119. Sethi S, Madden BJ, Debiec H, Charlesworth MC, Gross L, Ravindran branous nephropathy (IMN). Autoimmun Rev 15: 146–154, 2016
A, et al.: Exostosin 1/exostosin 2-associated membranous nephropathy. PubMed
J Am Soc Nephrol 30: 1123–1136, 2019 PubMed 137. Seitz-Polski B, Dolla G, Payre C, Girard CA, Polidori J, Zorzi K, et al.:
120. Sethi S, Madden B, Casal Moura M, Nasr SH, Klomjit N, Gross L, Epitope spreading of autoantibody response to PLA2R associates with
et al.: Hematopoietic stem cell transplant-membranous nephropathy is poor prognosis in membranous nephropathy. J Am Soc Nephrol 27:
associated with protocadherin FAT1. J Am Soc Nephrol 33: 1033–1044, 1517–1533, 2016 PubMed
2022 PubMed 138. Seitz-Polski B, Debiec H, Rousseau A, Dahan K, Zaghrini C, Payre C,
121. Caza TN, Hassen SI, Kuperman M, Sharma SG, Dvanajscak Z, Arthur et al.: Phospholipase A2 receptor 1 epitope spreading at baseline
J, et al.: Neural cell adhesion molecule 1 is a novel autoantigen in mem- predicts reduced likelihood of remission of membranous nephropathy.
branous lupus nephritis. Kidney Int 100: 171–181, 2021 PubMed J Am Soc Nephrol 29: 401–408, 2018 PubMed
122. Hoxha E, Kneißler U, Stege G, Zahner G, Thiele I, Panzer U, et al.: 139. Reinhard L, Zahner G, Menzel S, Koch-Nolte F, Stahl RAK, Hoxha E:
Enhanced expression of the M-type phospholipase A2 receptor in glo- Clinical relevance of domain-specific phospholipase A2 receptor 1 anti-
meruli correlates with serum receptor antibodies in primary membra- body levels in patients with membranous nephropathy. J Am Soc
nous nephropathy. Kidney Int 82: 797–804, 2012 PubMed Nephrol 31: 197–207, 2020 PubMed
123. Bobart SA, De Vriese AS, Pawar AS, Zand L, Sethi S, Giesen C, et al.: 140. Beck LH Jr, Fervenza FC, Beck DM, Bonegio RG, Malik FA, Erickson
Noninvasive diagnosis of primary membranous nephropathy using SB, et al.: Rituximab-induced depletion of anti-PLA2R autoantibodies
phospholipase A2 receptor antibodies. Kidney Int 95: 429–438, 2019 predicts response in membranous nephropathy. J Am Soc Nephrol 22:
PubMed 1543–1550, 2011 PubMed
124. Bobart SA, Han H, Tehranian S, De Vriese AS, Roman JCL, Sethi S, 141. Medrano AS, Escalante EJ, Caceres CC, Pamplona IA, Allende MT,
et al.: Noninvasive diagnosis of PLA2R-associated membranous Terrades NR, et al.: Prognostic value of the dynamics of M-type phos-
nephropathy: A validation study. Clin J Am Soc Nephrol 16: 1833– pholipase A2 receptor antibody titers in patients with idiopathic mem-
1839, 2021 PubMed branous nephropathy treated with two different immunosuppression
125. Qin W, Beck LH Jr, Zeng C, Chen Z, Li S, Zuo K, et al.: Anti-phos- regimens. Biomarkers 20: 77–83, 2015 PubMed
pholipase A2 receptor antibody in membranous nephropathy. J Am Soc 142. Ruggenenti P, Debiec H, Ruggiero B, Chianca A, Pelle T, Gaspari F,
Nephrol 22: 1137–1143, 2011 PubMed et al.: Anti-phospholipase A2 receptor antibody titer predicts post-
Nephrology Self-Assessment Program - Vol 21, No 5, December 2022 351

rituximab outcome of membranous nephropathy. J Am Soc Nephrol 26: nephropathy with the nephrotic syndrome. Nephrol Dial Transplant 21:
2545–2558, 2015 PubMed 3127–3132, 2006 PubMed
143. De Vriese AS, Glassock RJ, Nath KA, Sethi S, Fervenza FC: A proposal 155. Praga M, Barrio V, Juarez GF, Lu~ no J; Grupo Espa~ nol de Estudio de la
for a serology-based approach to membranous nephropathy. J Am Soc Nefropatıa Membranosa: Tacrolimus monotherapy in membranous nephrop-
Nephrol 28: 421–430, 2017 PubMed athy: A randomized controlled trial. Kidney Int 71: 924–930, 2007 PubMed
144. Ren S, Wu C, Zhang Y, Wang AY, Li G, Wang L, et al.: An update on 156. Ramachandran R, Hn HK, Kumar V, Nada R, Yadav AK, Goyal A,
clinical significance of use of THSD7A in diagnosing idiopathic et al.: Tacrolimus combined with corticosteroids versus modified Ponti-
membranous nephropathy: A systematic review and meta-analysis of celli regimen in treatment of idiopathic membranous nephropathy:
THSD7A in IMN. Ren Fail 40: 306–313, 2018 PubMed Randomized control trial. Nephrology (Carlton) 21: 139–146, 2016
145. Hoxha E, Beck LH Jr, Wiech T, Tomas NM, Probst C, Mindorf S, et al.: PubMed
An indirect immunofluorescence method facilitates detection of throm- 157. Caro J, Gutierrez-Solıs E, Rojas-Rivera J, Agraz I, Ramos N, Rabasco C,
bospondin type 1 domain-containing 7A-specific antibodies in membra- et al.; Grupo de Estudio de las Enfermedades Glomerulares de la Sociedad
nous nephropathy. J Am Soc Nephrol 28: 520–531, 2017 PubMed Espa~ nola de Nefrologıa (GLOSEN): Predictors of response and relapse in
146. Fila M, Debiec H, Perrochia H, Djouadi N, Verpont MC, Buob D, et al.: patients with idiopathic membranous nephropathy treated with tacroli-
Recurrence of anti-semaphorin 3B-mediated membranous nephropathy mus. Nephrol Dial Transplant 30: 467–474, 2015 PubMed
after kidney transplantation. J Am Soc Nephrol 33: 503–509, 2022 158. Remuzzi G, Chiurchiu C, Abbate M, Brusegan V, Bontempelli M,
PubMed Ruggenenti P: Rituximab for idiopathic membranous nephropathy.
147. Ravindran A, Casal Moura M, Fervenza FC, Nasr SH, Alexander MP, Lancet 360: 923–924, 2002 PubMed
Fidler ME, et al.: In patients with membranous lupus nephritis, 159. Fervenza FC, Abraham RS, Erickson SB, Irazabal MV, Eirin A, Specks
exostosin-positivity and exostosin-negativity represent two different phe- U, et al.; Mayo Nephrology Collaborative Group: Rituximab therapy in
notypes. J Am Soc Nephrol 32: 695–706, 2021 PubMed idiopathic membranous nephropathy: A 2-year study. Clin J Am Soc
148. Wang C, Liu Y, Zhang M, Yang F, Xu F, Shi S, et al.: Glomerular exo- Nephrol 5: 2188–2198, 2010 PubMed
stosin as a subtype and activity marker of class 5 lupus nephritis. Clin 160. Dahan K, Debiec H, Plaisier E, Cachanado M, Rousseau A, Wakselman L,
J Am Soc Nephrol 17: 986–993, 2022 PubMed et al.; GEMRITUX Study Group: Rituximab for severe membranous
149. Ruggenenti P, Cravedi P, Chianca A, Perna A, Ruggiero B, Gaspari F, nephropathy: A 6-month trial with extended follow-up. J Am Soc Nephrol
et al.: Rituximab in idiopathic membranous nephropathy. J Am Soc 28: 348–358, 2017 PubMed
Nephrol 23: 1416–1425, 2012 PubMed 161. Fervenza FC, Appel GB, Barbour SJ, Rovin BH, Lafayette RA, Aslam
150. Jha V, Ganguli A, Saha TK, Kohli HS, Sud K, Gupta KL, et al.: A ran- N, et al.; MENTOR Investigators: Rituximab or cyclosporine in the
domized, controlled trial of steroids and cyclophosphamide in adults treatment of membranous nephropathy. N Engl J Med 381: 36–46,
with nephrotic syndrome caused by idiopathic membranous nephropa- 2019 PubMed
thy. J Am Soc Nephrol 18: 1899–1904, 2007 PubMed 162. Fernandez-Juarez G, Rojas-Rivera J, Logt AV, Justino J, Sevillano A, Cara-
151. Ponticelli C, Zucchelli P, Passerini P, Cesana B, Locatelli F, Pasquali S, vaca-Fontan F, et al.; STARMEN Investigators: The STARMEN trial indi-
et al.: A 10-year follow-up of a randomized study with methylpredniso- cates that alternating treatment with corticosteroids and cyclophosphamide
lone and chlorambucil in membranous nephropathy. Kidney Int 48: is superior to sequential treatment with tacrolimus and rituximab in pri-
1600–1604, 1995 PubMed mary membranous nephropathy. Kidney Int 99: 986–998, 2021 PubMed
152. Cattran DC, Greenwood C, Ritchie S, Bernstein K, Churchill DN, 163. Scolari F, Delbarba E, Santoro D, Gesualdo L, Pani A, Dallera N, et al.;
Clark WF, et al.; Canadian Glomerulonephritis Study Group: A con- RI-CYCLO Investigators: Rituximab or cyclophosphamide in the treat-
trolled trial of cyclosporine in patients with progressive membranous ment of membranous nephropathy: The RI-CYCLO randomized trial.
nephropathy. Kidney Int 47: 1130–1135, 1995 PubMed J Am Soc Nephrol 32: 972–982, 2021 PubMed
153. Cattran DC, Appel GB, Hebert LA, Hunsicker LG, Pohl MA, Hoy WE, 164. Podesta MA, Ruggiero B, Remuzzi G, Ruggenenti P: Ofatumumab for
et al.; North America Nephrotic Syndrome Study Group: Cyclosporine multirelapsing membranous nephropathy complicated by rituximab-
in patients with steroid-resistant membranous nephropathy: A random- induced serum-sickness. BMJ Case Rep 13: e232896, 2020 PubMed
ized trial. Kidney Int 59: 1484–1490, 2001 PubMed 165. Sethi S, Kumar S, Lim K, Jordan SC: Obinutuzumab is effective for the
154. Alexopoulos E, Papagianni A, Tsamelashvili M, Leontsini M, Memmos D: treatment of refractory membranous nephropathy. Kidney Int Rep 5:
Induction and long-term treatment with cyclosporine in membranous 1515–1518, 2020 PubMed

You might also like