Vous êtes sur la page 1sur 11

CLINICAL RESEARCH www.jasn.

org

Rituximab for Severe Membranous Nephropathy:


A 6-Month Trial with Extended Follow-Up
Karine Dahan,* Hanna Debiec,†‡ Emmanuelle Plaisier,*†‡ Marine Cachanado,§
Alexandra Rousseau,§ Laura Wakselman,§ Pierre-Antoine Michel,* Fabrice Mihout,*
Bertrand Dussol,| Marie Matignon,¶ Christiane Mousson,** Tabassome Simon,§ and
Pierre Ronco,*†‡ on behalf of the GEMRITUX Study Group
*Department of Nephrology and Dialysis, Assistance Publique Hôpitaux de Paris, Hôpital Tenon, Paris, France;

Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Paris, France; ‡Institut National de la Santé et de la
Recherche Médicale, Unité Mixte de Recherche 1155, Paris, France; §Department of Clinical Pharmacology and Unité
de Recherche Clinique, Assistance Publique Hôpitaux de Paris, Hôpital Saint Antoine, Paris, France; |Department of
Nephrology and Transplantation, Assistance Publique-Hôpitaux de Marseille, Hôpital de la Timone, Marseille, France;

Department of Nephrology and Transplantation, Assistance Publique Hôpitaux de Paris, Hôpital Henri Mondor,
Creteil, France; and **Department of Nephrology and Transplantation, Centre Hospitalier Universitaire, Dijon, France

ABSTRACT
Randomized trials of rituximab in primary membranous nephropathy (PMN) have not been conducted. We
undertook a multicenter, randomized, controlled trial at 31 French hospitals (NCT01508468). Patients with
biopsy-proven PMN and nephrotic syndrome after 6 months of nonimmunosuppressive antiproteinuric treat-
ment (NIAT) were randomly assigned to 6-month therapy with NIAT and 375 mg/m2 intravenous rituximab on
days 1 and 8 (n=37) or NIAT alone (n=38). Median times to last follow-up were 17.0 (interquartile range, 12.5–
24.0) months and 17.0 (interquartile range, 13.0–23.0) months in NIAT-rituximab and NIAT groups, respec-
tively. Primary outcome was a combined end point of complete or partial remission of proteinuria at 6 months.
At month 6, 13 (35.1%; 95% confidence interval [95% CI], 19.7 to 50.5) patients in the NIAT-rituximab group
and eight (21.1%; 95% CI, 8.1 to 34.0) patients in the NIAT group achieved remission (P=0.21). Rates of
antiphospholipase A2 receptor antibody (anti–PLA2R-Ab) depletion in NIAT-rituximab and NIAT groups were
14 of 25 (56%) and one of 23 (4.3%) patients at month 3 (P,0.001) and 13 of 26 (50%) and three of 25 (12%)
patients at month 6 (P=0.004), respectively. Eight serious adverse events occurred in each group. During the
observational phase, remission rates before change of assigned treatment were 24 of 37 (64.9%) and 13 of 38
(34.2%) patients in NIAT-rituximab and NIAT groups, respectively (P,0.01). Positive effect of rituximab on
proteinuria remission occurred after 6 months. These data suggest that PLA2R-Ab levels are early markers of
rituximab effect and that addition of rituximab to NIAT does not affect safety.

J Am Soc Nephrol 28: ccc–ccc, 2016. doi: 10.1681/ASN.2016040449

Membranous nephropathy (MN) accounts for identification of the podocyte antigen phospholi-
about 20% of cases of nephrotic syndrome in adults pase A2 receptor (PLA2R) as the target of circulating
and is the leading glomerulopathy recurring after
kidney transplantation.1 Thickening of glomerular
capillary walls results from subepithelial formation Received April 19, 2016. Accepted May 22, 2016.

of immune deposits containing IgG, the membrane Published online ahead of print. Publication date available at
attack complex of complement, which is the major www.jasn.org.
mediator of proteinuria, and antigens. Primary Correspondence: Dr. Karine Dahan or Prof. Pierre Ronco, Tenon
forms of MN, improperly called primary membra- Hospital, 4 rue de la Chine, F-75020 Paris, France. Email: karine.
dahan@aphp.fr or pierre.ronco@upmc.fr
nous nephropathy (PMN), represent 70%–80%
of all patients. A major breakthrough was the Copyright © 2016 by the American Society of Nephrology

J Am Soc Nephrol 28: ccc–ccc, 2016 ISSN : 1046-6673/2801-ccc 1


CLINICAL RESEARCH www.jasn.org

antibodies in about 70% of PMN, which confirmed that the (Figure 1). Three patients withdrew their consent before
disease was autoimmune in nature.2 randomization. Thirty-nine patients were assigned to the
The optimal treatment of patients with PMN is still a matter nonimmunosuppressive antiproteinuric treatment (NIAT)-
of debate3,4; 30%–40% of affected patients will undergo spon- rituximab group, and 38 patients were assigned to the NIAT
taneous, usually partial remission usually within 1 year from only group. Thirty-seven patients in the NIAT-rituximab
disease onset, whereas about one third will progress to group and 38 in the NIAT group received the assigned treat-
ESRD. 5,6 Treatments with corticosteroids and alkylating ment. Baseline characteristics in the two groups were similar
agents significantly increase the rates of remission and slow (Table 1).
renal function loss in patients with persistent nephrotic syn-
drome.7–11 Calcineurin inhibitors induce remission in a ma- Primary End Point
jority of patients, but relapse rates exceed 50%; also, renal The 6-month trial failed to achieve the primary end point.
toxicity is a concern.10,12–14 The latest Kidney Disease Improv- Thirteen patients (35.1%; 95% confidence interval [95% CI],
ing Global Outcomes (KDIGO) guidelines
restricted the indication of alkylating
agents to patients at high risk of progres-
sion and considered calcineurin inhibitors
as an alternative therapy.15 In patients with
even more restricted indications of alkylat-
ing agents, the rate of serious adverse
events (SAEs), particularly malignancy,
was higher in patients who received long-
term immunosuppression than in those
with supportive therapy.16
Both the evidence that B cells play a key
role in the pathogenesis of PMN and drug
toxicity led to target B cells with rituxi-
mab.17 Rituximab induced remission of
nephrotic syndrome in 60%–80% of the
patients with long-lasting proteinuria, de-
spite blockade of the renin-angiotensin
system,18–21 and in patients who had pre-
viously failed other treatments. Reduction
of PLA2R-Ab titer preceded remission of
proteinuria by several months, which
suggested a causal relationship.22,23 A pre-
vious study showed that a B cell–driven ap-
proach with only one or two infusions of
rituximab of 375 mg/m2 per week could
allow reducing cost in comparison with
the standard four weekly infusions.24
Because of the lack of randomized, con-
trolled trials (RCTs) using rituximab and the
high rate of spontaneous remission, this trial
was designed to evaluate the efficacy of
rituximab given to all patients at a standard
dose (375 mg/m2) in two infusions added to
supportive therapy compared with support-
ive therapy alone in patients with persistent
nephrotic syndrome.
Figure 1. Flow chart of the trial. The flow chart shows that premature discontinuation
occurred in five patients within 3 months after inclusion: (1) two remissions at day 1 or
RESULTS inclusion; (2) one in NIAT for ,6 months; (3) one lost to follow-up and one diagnosed
with a pulmonary neoplasia; and (4) three treatment shifts between 3 and 6 months:
Between January 17, 2012, and July 3, 2014, two received rituximab or steroids because of deterioration of clinical condition and
80 patients were enrolled in the RCT phase one was referred to another center.

2 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org CLINICAL RESEARCH

Table 1. Baseline characteristics


Characteristic NIAT-Rituximab Group, n=37 NIAT Group, n=38 Total, n=75
Age, yr 53.0 (42.0–63.0) 58.5 (43.0–64.0) 56 (42.0–64.0)
Men, no. (%) 28 (75.7) 24 (63.2) 52 (69.3)
Weight, kg 76.0 (70.0–85.0) 76.5 (67.0–85.0) 76.0 (67.0–85.0)
BP, mmHg
Systolic 124 (110–140) 125 (117–140) 125 (115–140)
Diastolic 77 (68–82) 76 (70–81) 76 (70–81)
Creatinine, mmol/L 98.1 (73.4–122.9) 91.1 (74.3–122.0) 93.8 (76.9–122.9)
eGFR, ml/min per 1.73 m2 66.7 (55.4–82.5) 72.7 (58.1–88.6) 68.6 (55.4–88.6)
Protein-to-creatinine ratio, mg/g 7680.0 (4584.3–10,399.0) 7195.1 (5363.1–8965.1) 7363.2 (4702.9–9735.0)
Albumin level, g/L 22 (18–25) 22 (20–26) 22 (19–26)
Median time since biopsy-proven diagnosis, mo 8 (6–13) 8 (6–11) 8 (6–13)
PLA2R-Ab–positive patients (ELISA), no. (%) 27 (73.0) 28 (73.7) 55 (73.3)
PLA2R-Ab titer (ELISA), RU/mla 40.5 (0.0–275.5) 43.3 (0.0–457.5) 40.5 (0.0–440.9)
Diuretics, no. (%) 32 (86.5) 32 (84.2) 64 (85.3)
ACE inhibitor and/or ARB, no. (%)
ACE inhibitor 16 (43.2) 14 (38.9) 30 (41.1)
ARB 12 (32.4) 8 (22.2) 20 (27.4)
ACE inhibitor and ARB 9 (24.3) 14 (38.9) 23 (31.51)
Statins, no. (%) 31 (83.8) 26 (68.4) 57 (76.0)
Data are shown as n (%) or median (IQR). eGFR was calculated according to the MDRD equation. ACE, angiotensin-converting enzyme; ARB, angiotensin 2 receptor
blocker.
a
Median and IQR of PLA2R-Ab titer in all patients with and without PLA2R-Ab.

19.7% to 50.5%) in the NIAT-rituximab group and eight observed at month 3 (0.0; IQR, 0.0–60.5 RU/ml; P,0.001) and
patients (21.1%; 95% CI, 8.1% to 34.0%) in the NIAT group month 6 (8.3; IQR, 0.0–73.5 RU/ml; P,0.001) compared with
achieved proteinuria remission at month 6 after randomiza- baseline (102.5; IQR, 36.1–672.5 RU/ml) in the NIAT-
tion (P=0.21; odds ratio [OR], 2.0; 95% CI, 0.7 to 5.7) (Table rituximab group and only at month 6 (62.9; IQR, 16.6–49.3
2). Results were not sensitive to missing data replacement. RU/ml versus 199.5; IQR, 24.2–491.4 RU/ml at baseline;
P=0.02) in the NIAT alone group. Percentage decrease of
Secondary End Points PLA2R-Ab titer was significantly higher in the NIAT-
Protein-to-creatinine ratio decreased similarly in both groups rituximab group at months 3 and 6 (Figure 2C).
at months 3 and 6 (Figure 2A, Table 2). Percentage increase of Complete immunologic remission (full PLA2R-Ab deple-
serum albumin was significantly higher at months 3 and 6 in tion) was observed in the NIAT-rituximab group in 14 of 25
the NIAT-rituximab group (Figure 2B). Serum creatinine and (56%) and 13 of 26 (50%) patients at months 3 and 6,
eGFR by Modification of Diet in Renal Disease (MDRD) for- respectively, compared with one of 23 (4%; P,0.001) and
mula (Table 2), serum triglycerides, total and LDL cholesterol, three of 25 (12%; P=0.004) patients, respectively, in the
body weight, and need for diuretic therapy did not differ at NIAT group (Table 2). Of the 14 rituximab-treated patients
months 3 and 6 between the two groups (Supplemental Table 1). that were antibody depleted at month 3, six (43%) patients
PLA2R-Ab was detected in 27 (73%) and 28 (74%) patients subsequently achieved the primary end point compared with
at baseline in the NIAT-rituximab and the NIAT groups, only two of 11 patients (18%) without antibody depletion.
respectively (Table 1). As early as month 3, rates of PLA2R- PLA2R-Ab level ,275 RU/ml at baseline was associated with
Ab–positive patients (31% versus 83%; P,0.001) and PLA2R- the primary end point (OR, 4.3; 95% CI, 1.1 to 17.3; P=0.04),
Ab titers (0.0; interquartile range [IQR], 0.0–49.1 RU/ml and this was independent from treatment group, age, sex,
versus 54.6; IQR, 16.5–278.4 RU/ml; P,0.001) were lower baseline proteinuria, serum albumin, and creatinine (Supple-
in the NIAT-rituximab group than in the NIAT alone, respec- mental Table 2).
tively (Table 2). No further decrease in the rate of PLA2R-Ab– PLA2R-Ab was also measured at a very early time point (day
positive patients was observed between months 3 and 6, and 8). Rates of PLA2R-Ab positivity and PLA2R-Ab titer in the
the difference between PLA2R-Ab titer at months 3 and 6 was whole population and PLA2R-Ab titer in the subset of patients
0.0 (IQR, 0.0–19.8) in the NIAT-rituximab group. No change who were positive at baseline were similar in both treatment
in the rates of PLA2R-Ab–positive patients and PLA2R-Ab ti- groups (Table 2). Among the eight patients of the NIAT-
ters occurred between baseline, and months 3 and 6 in the rituximab group who were PLA2R positive at baseline and
NIAT group (Table 2). achieved remission at 6 months, two were antibody depleted
In the subgroup of patients who were initially positive for at day 8, and two had marked reductions (78% and 42%) of
PLA2R-Ab, significant decreases of the titers of PLA2R-Ab were antibody titer.

J Am Soc Nephrol 28: ccc–ccc, 2016 Rituximab for Membranous Nephropathy 3


CLINICAL RESEARCH www.jasn.org

Table 2. Efficacy outcome variables


Variable NIAT-Rituximab Group, n=37 NIAT Group, n=38 P Value
Remission, complete and partiala 13 (35.1; 19.7 to 50.5) 8 (21.1; 8.1 to 34.0) 0.21
Protein-to-creatinine ratio, mg/g
Baseline 7680.0 (4584.3–10,399.0) 7195.1 (5363.1–8965.1)
3 mo 4814.4 (3205.5–7398.6) 4832.1 (2424.9–7911.9) 0.94b
6 mo 3531.2 (1796.6–6469.4) 5265.8 (2500.1–7690.7) 0.18b
Serum albumin level, g/L
Baseline 22 (18–25) 22 (20–26)
3 mo 27 (21–31) 23 (19–27) 0.10b
6 mo 30 (26–34) 24 (20–29) 0.029b
Serum creatinine, mmol/L
Baseline 98.1 (82.2–122.9) 91.1 (74.3–122.0)
3 mo 94.6 (78.7–114.0) 100.8 (81.3–115.8) 0.88b
6 mo 94.6 (75.1–130.8) 97.2 (76.0–126.4) 0.67b
eGFR, ml/min per 1.73 m2
Baseline 66.7 (55.4–82.5) 72.7 (58.1–88.6)
3 mo 66.7 (57.2–87.1) 68.9 (45.7–89.7) 0.95b
6 mo 65.6 (51.0–89.0) 72.5 (52.4–89.7) 0.75b
PLA2R-Ab–positive patients, ELISA
Baseline 27 (73.0) 28 (73.7)
Day 8 18 (52.9) 17 (68.0) 0.25c
3 mo 11 (31.4) 25 (83.3) ,0.001c
6 mo 13 (36.1) 24 (75.0) 0.001c
PLA2R-Ab–depleted patients
3 mo 14/25 (56.0) 1/23 (4.3) ,0.001b
6 mo 13/26 (50.0) 3/25 (12.0) 0.004b
PLA2R-Ab titer (all patients), RU/ml
Baseline 40.5 (0.0–275.5) 43.3 (0.0–457.5)
Day 8 27.1 (0.0–126.1) 65.5 (0.0–345.5) 0.24c
3 mo 0.0 (0.0–49.1) 54.6 (16.5–278.4) ,0.001c
6 mo 0.0 (0.0–34.0) 45.7 (7.6–262.2) 0.002c
PLA2R-Ab titer (positive patients),d RU/ml
Baseline 102.5 (36.1–672.5) 199.5 (24.2–491.4)
Day 8 63.2 (12.9–382.0) 163.5 (34.7–438.5) 0.41c
3 mo 0.0 (0.0–60.5) 77.5 (30.3–325.9) 0.003c
6 mo 8.3 (0.0–73.5) 62.9 (16.6–449.3) 0.01c
Post hoc composite end point at 6 mo 15 (40.5; 24.7 to 56.4) 5 (13.2; 2.4 to 23.9) ,0.01
CD19, per mm3e
3 mo 11 (2.0–22.0) NA
6 mo 61 (34.0–100) NA
Data are shown as n (%), n (%; 95% CI), or median (IQR). eGFR was calculated according to the MDRD equation. NA, not available.
a
Complete and partial remissions were defined according to 2012 KDIGO criteria on the basis of proteinuria; composite end point was defined as reduction of
proteinuria .50% and increase of serum albumin .30%.
b
P value ,0.03 indicates statistical significance (Bonferroni correction).
c
P value ,0.02 indicates statistical significance (Bonferroni correction).
d
Patients with at least one positive detection of PLA2R-Ab at any time.
e
Normal range (100–500/mm3).

Twenty patients had undetectable PLA2R-Ab at baseline (10 domain 7A antibody (anti–THSD7A-Ab). The first patient
in the NIAT-rituximab group and 10 in the NIAT group). (1/100 dilution at baseline) received NIAT-rituximab and
However, three patients in the NIAT group later developed achieved remission at month 6, with sustained antibody de-
PLA2R-Ab and were considered as having PLA2R-related pletion from month 3. The second patient was treated with
PMN. At baseline, no statistical difference in age, protein-to- NIAT alone and achieved partial remission at month 6 but
creatinine ratio, serum albumin, and eGFR was seen according relapsed at 1 year. In this patient, THSD7A-Ab remained de-
to presence (n=55 patients) or absence (n=20) of PLA2R-Ab. tectable at low level (1/10) at any time.
The effect of rituximab on proteinuria remission did not differ CD19+ B cells remained below normal range (100–500/mm3)
according to the serologic status at baseline. Two PLA2R-Ab– throughout the observation period in the NIAT-rituximab
negative patients were positive for antithrombospondin group. Median CD19+ B cell count was 11/mm 3 (IQR,

4 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org CLINICAL RESEARCH

Post Hoc Composite End Point


In a post hoc analysis that includes reduction of proteinuria
.50% and increase of serum albumin level .30% at month 6,
15 (41%) patients in the NIAT-rituximab group and five
(13%) patients in the NIAT group achieved the composite
end point at month 6 after randomization (OR, 0.22; 95%
CI, 0.07 to 0.70; P,0.01) (Table 2).

Post–RCT Observational Period


Median duration from inclusion to last follow-up was 17.0
(IQR, 12.5–24.0) months and 17.0 (IQR, 13.0–23.0) months
in the NIAT-rituximab and the NIAT groups, respectively. The
rates of KDIGO remission occurring without modification of
initial immunosuppressive treatment (NIAT-rituximab) or
introduction of an immunosuppressive treatment (NIAT)
were 24 of 37 (64.9%) and 13 of 38 (34.2%) in the NIAT-
rituximab and the NIAT groups, respectively (OR, 3.5; 95%
CI, 1.7 to 9.2; P,0.01) (Table 3). Numbers of complete re-
mission were seven of 37 and one of 38 in the NIAT-rituximab
and the NIAT groups, respectively (P=0.03). Median times to
remission were 7.0 (IQR, 5.5–10.5) months (n=24) and 7.0
(4.0–13.0) months (n=13) in the NIAT-rituximab and the
NIAT groups, respectively. Protein-to-creatinine ratio was
lower in the NIAT-rituximab group (2194.8; IQR, 1309.8–
5310.0 mg/g) than in the NIAT group (4701.1; IQR, 2027.8–
8265.3; P=0.02), whereas serum albumin level was higher (32;
IQR, 26–35 g/L versus 27; 20–30 g/L, respectively; P=0.03).
Serum creatinine and eGFR by MDRD formula did not differ
between the two groups (Table 3). In multivariate analyses,
KDIGO remission was associated with PLA2R-Ab level ,275
RU/ml at baseline (OR, 3.5; 95% CI, 1.1 to 10.7; P=0.03), and
this was independent from treatment group, age, sex, baseline
proteinuria, serum albumin, and creatinine (Supplemental
Table 3). KDIGO remission was also associated with compos-
ite end point at month 6 (OR, 30.1; 95% CI, 3.9 to 262.8;
P=0.001), regardless of treatment group. In the NIAT-
rituximab group, CD19 counts at months 3 and 6 were not
associated with remission.
Figure 2. The secondary end points are expressed as percentage
changes in proteinuria, serum albumin, and PLA2R-Ab with time. SAEs
Mean6SEM percentage changes from baseline in (A) proteinuria, Number of SAEs was comparable in both groups (Table 4).
(B) serum albumin, and (C) anti–PLA2R-Ab levels. Please note that Only one SAE was related to NIAT-rituximab treatment in a
C shows percentage changes of PLA2R antibodies in the subset patient who developed prostatitis with favorable outcome. In
of patients who had PLA2R-Ab at baseline. Bonferroni correction the rituximab group, no leukopenia was observed. Patients
was applied; P value ,0.02 indicates statistical significance. NIAT received a premedication protocol of 100 mg solumedrol, 1
is shown by blue lines, and NIAT-rituximab is shown by red lines. g paracetamol, and 5 mg dexchlorpheniramine; no allergic
*P,0.02; **P,0.001; ***P,0.001. reactions were observed.

2.0–22.0) at month 3 and 61/mm3 (IQR, 34.0–100) at month 6 DISCUSSION


(Table 2). Among PLA2R-Ab–positive patients at baseline,
there was no difference in CD19 count between patients In this randomized study, we analyzed the effect of rituximab
who were PLA2R-Ab depleted and those who were not at combined with NIAT in patients with PMN and severe
month 3 (P=0.76; n=23 patients) and month 6 (P=0.89; nephrotic syndrome who had resisted maximally tolerated
n=24 patients). antiproteinuric therapy. The RCTshowed that, compared with

J Am Soc Nephrol 28: ccc–ccc, 2016 Rituximab for Membranous Nephropathy 5


CLINICAL RESEARCH www.jasn.org

Table 3. Results of efficacy analysis at last follow-up


End Point NIAT-Rituximab Group, n=37 NIAT Group, n=38 P Value
Remission, complete and partiala 24 (64.9; 49.5 to 80.2) 13 (34.2; 19.1 to 49.3) ,0.01
Protein-to-creatinine ratio, mg/g 2194.8 (1309.8–5310.0) 4701.1 (2027.8–8265.3) 0.02
Serum albumin, g/L 32 (26–35) 27 (20–30) 0.03
Serum creatinine, mmol/L 101 (87–135) 97.2 (78.5–133.5) 0.50
eGFR, ml/min per 1.73 m2 61.1 (48.7–83.4) 73.1 (50.4–90.5) 0.48
Data are shown as n (%; 95% CI) or median (IQR). Data were recorded before any potential modification of treatment assigned at randomization (modification of
initial immunosuppressive treatment in the NIAT-rituximab group or addition of any immunosuppressive treatment in the NIAT group).
a
Complete and partial remissions were defined according to 2012 KDIGO criteria on the basis of proteinuria.

Table 4. SAEs according to treatment group on NIAT for .6 months. Because no major
complication occurred in the NIAT group,
NIAT-Rituximab NIAT Group,
SAEs P Value the risk taken was acceptable; however, af-
Group, n=37 n=38
ter 6 months, PLA2R-Ab–positive patients
No. of events 0.87
had a markedly higher antibody titer in the
0 31 33
NIAT group than in the NIAT-rituximab
1 5 4
$2 1 1
group, and it is uncertain whether a delay
Event details by 1 year in the NIAT group would affect
Acute renal failure a
0 2 any future response to immunosuppressive
Infection agents or rituximab therapy. Alternatively,
Prostatitis 1 0 we could have set the end point at 12
Pleural effusiona 0 1 months with prespecified measures in the
Cardiac and vascular disorders most aggressive forms for the patients in
Myocardial infarction 1 1 the NIAT group. However, this protocol
Critical limb ischemia 0 1 would have assessed a global treatment
Mesenteric ischemiab 1 0
strategy (NIATwith or without retreatment
Carotid endarteriectomyb 1 0
b and NIAT with rituximab with or without
Aortoiliac femoral bypass 1
Cancer a
0 1
retreatment) and not only the efficacy of
Others rituximab added to supportive therapy.
Edema 1 1 We, therefore, opted for a pragmatic ap-
Pain and fever 1 0 proach, with a 6-month period of RCT fol-
Diarrhea 1 0 lowed by an observational phase.
Asthma 0 1 This RCT failed to reach the primary end
Data are shown as n. point. The lack of effect of NIAT-rituximab
a
These SAEs occurred in the same patient.
b on the rate of proteinuria remission at 6
These SAEs occurred in the same patient.
months has several explanations: (1) the
high rate of remission (21%) in the NIAT
NIAT alone, addition of two infusions of rituximab to NIAT group; (2) the lower rate of remission (35%) in the NIAT-
decreased PLA2R-Ab as early as month 3 and was associated rituximab group than we expected, because sample size was
with a higher percentage increase of serum albumin at months calculated from initial studies on rituximab,17,18,25 which
3 and 6. However, the effect of this combined treatment on the probably overestimated the rate of remission in the NIAT-
rate of proteinuria remission (primary end point) was not rituximab group and led to a lack of power; (3) the short
observed during the RCT but was delayed to the post–RCT duration of the RCT phase; and (4) the fact that proteinuria
observational period (median time to remission of 7 months). is a delayed marker of treatment effect.20,21,23
The trial, thus, provides new biomarkers of early treatment However, we did observe an effect of rituximab on serum
response. albumin variation from baseline (increase) and PLA2R-Ab
We compared NIAT-rituximab with NIAT alone, because levels as early as month 3, which was confirmed at month 6. The
there was no evidence-based proof of the efficacy of rituximab increase from baseline of serum albumin contrasting with
in PMN, although several nonrandomized studies suggested persisting high–level proteinuria at month 6 in the NIAT-
that rituximab was efficient18–21; the possibility of bias linked rituximab group might be related to decreased tubular reab-
to a high rate of late spontaneous remissions as confirmed in sorption of albumin when serum albumin increases26 and/or
this study called for a randomized trial. With this aim in mind, increased albumin anabolic rate in the liver resulting in in-
an ideal trial would have been a prolonged trial for .1 year. creasing protein load to the glomerulus, which would offset
However, we considered it unethical to maintain the patients the improving glomerular sieving function. 27 We, thus,

6 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org CLINICAL RESEARCH

considered a post hoc composite end point with the aim to point) and during the post–RCT observational phase without
define early clinical criteria of response to rituximab, and it modification of treatment assigned at randomization, regard-
was a reduction of proteinuria .50% and an increase of serum less of treatment group and other adjustment variables. Our
albumin level .30% at month 6. A significantly higher num- results also suggest that THSD7A-Ab may be useful for the
ber of patients treated with NIAT-rituximab compared with monitoring of patients with MN. However, our univariate and
NIATreached this composite end point at month 6. Moreover, multivariate analyses failed to identify classic predictors of
remission defined on composite end point at month 6 was long-term outcome and proteinuria remission, such as pro-
associated with proteinuria remission occurring at any time teinuria, serum creatinine, eGFR, serum albumin, age, and
before any change of initially assigned treatment. This com- sex. A possible reason is that we studied a relatively homoge-
posite end point might, therefore, better reflect early renal neous population after a 6-month run–in period of maximally
outcome, although it should be validated in additional studies. tolerated conservative therapy. This discrepant observation
We continued to follow the patients during a post–RCT gives even more importance to PLA2R-Ab as a predictor of
observational phase. The suspected beneficial clinical effect proteinuria remission in agreement with the autoimmune na-
of rituximab at month 6 was confirmed by the data of the ture of the disease. The finding that PLA2R-Ab positivity and
observational phase, which were recorded before any potential titer tended to decrease as early as 8 days in the NIAT-
modification of treatment assigned at randomization. Pro- rituximab group was somewhat unexpected given that the
teinuria remission rate was substantially higher in the NIAT- half-life of Igs is about 3 weeks, but it confirmed previous ob-
rituximab group than in the NIAT alone group (64.9% versus servations by Hoxha et al.30 This suggests that PLA2R-Ab might
37.5%), with proteinuria dropping to a much lower level in be a very early biomarker of rituximab efficacy, although this
the NIAT-rituximab group. In the patients treated with NIAT- has to be confirmed in additional prospective studies.
rituximab, proteinuria remission rate and median time to re- Second, this trial shows that B cell counts in rituximab-
mission (7 months) during the follow-up study were similar to treated subjects do not predict proteinuria remission and
those reported in previous nonrandomized series.20,21 confirms that PLA2R-Ab depletion rather than CD20+ de-
Remission rate was comparable with the one achieved with pletion achieved in all patients matters for prediction of
the Ponticelli protocol in the same time frame (50% at 6 rituximab response.23 It does not tell us, however, whether the
months8 and 32% within 1 year9) considering that, in those absence of immunologic remission at 3 months is because of
studies, patients were enrolled without a run-in period. It was lack of efficacy of rituximab or insufficient dosage or whether
lower than in patients treated with cyclosporin13 and tacrolimus14 patients without antibody depletion at 6 months should be
who had remission rates of 75% after 26 weeks and 58% after 6 reinfused or shifted to a new generation anti–CD20 antibody
months, respectively; however, baseline serum albumin was or another immunosuppressant.
higher by .0.5 g/dl than in our study, and these drugs are Third, we made the important observation that two
known to have an effect on glomerular hemodynamics, infusions of rituximab were not associated with an increased
with a high risk of relapse at discontinuation, and be associated risk of SAEs, which differs from all of the other current
with a clinically relevant nephrotoxicity. The ongoing Mem- immunosuppressive therapies for PMN.
branous Nephropathy Trial of Rituximab (ClinicalTrials.gov This trial has certain limitations. First, 11 patients among
no. NCT01180036) will hopefully show whether rituximab is the 24 patients who entered remission in the NIAT-rituximab
superior to cyclosporin in terms of proteinuria remission group reached the primary end point during the post–RCT
over a 24-month period. The high rate of spontaneous remis- observational study compared with only four in the NIAT
sion that may occur .1 year after disease onset in our study and group. However, the observational nature of the data does
the relatively low rate of remission with NIAT-rituximab, as not provide similar strength of evidence as those provided
with other immunosuppressive treatments, indicate that we by the randomized period. Second, one cannot exclude that
have not yet reached an optimal treatment in patients with some patients without circulating PLA2R-Ab at treatment
persisting nephrotic syndrome. It is difficult to extrapolate onset had PLA2R-related PMN. This question could be ad-
what would be the remission rate with rituximab only, because dressed by analysis of kidney biopsies,31 which was not possi-
all patients with persisting nephrotic syndrome are treated with ble in this multicentric trial. Third, the trial was not blinded.
NIAT according to the KDIGO recommendations. However, data assessors were blinded to treatment allocation,
This trial has several strengths. First, it is the first RCT in and SAEs were monitored by an independent organization.
patients with PMN with monitoring of PLA2R-Ab, detected in Fourth, the trial was too short to determine whether the re-
71% of the patients as in previous studies,28 and THSD7A- lapse rate was influenced by immunosuppressive treatment.
Ab. 29 Rituximab associated with NIAT reduced median Most of the remissions were partial. Because relapses of ne-
PLA2R-Ab titer as early as month 3 and induced complete phrotic syndrome and disease progression are more frequent
immunologic remission in 56% and 50% of the patients at in patients with partial remission, long-term studies with
months 3 and 6, respectively. Multivariate analyses showed rituximab are advocated.
that PLA2R-Ab,275 RU/ml at baseline was the only factor In conclusion, this trial shows that serum albumin and
associated with remission occurring at month 6 (primary end PLA2R-Ab levels are early markers of NIAT-rituximab efficacy,

J Am Soc Nephrol 28: ccc–ccc, 2016 Rituximab for Membranous Nephropathy 7


CLINICAL RESEARCH www.jasn.org

whereas the effect on proteinuria remission appears after 6 maximal tolerated dose of NIAT (angiotensin–converting enzyme
months. Addition of rituximab to NIAT has no effect on safety. inhibitors and/or angiotensin 2 receptor blockers, diuretics, and sta-
This first RCT is another step toward the use of rituximab as tin). Proteinuria was measured repeatedly before inclusion and treat-
first-line therapy in severe forms of PMN. It also suggests that ment assignment to confirm persistence of full–blown nephrotic
criteria for definition of remission should include serum syndrome. The eGFR by MDRD formula had to be .45 ml/min
albumin and PLA2R-Ab levels, particularly in trials where rapid per 1.73 m2. Exclusion criteria were secondary MN, pregnancy,
responses on drug efficacy and surrogate criteria are needed. breastfeeding, immunosuppressive treatment in the 3 preceding
months, and active infectious disease. Hepatitis B serology included
Hbs antigen and Hbs and Hbc antibodies. Patients with active hep-
CONCISE METHODS atitis B and those with past hepatitis B infection without anti-Hbs
antibodies were excluded. Four patients had previously received che-
Study Design motherapy according to the Ponticelli protocol: one in the NIAT-
Patients were enrolled at 31 hospital nephrology units throughout rituximab group had chemotherapy completed 13 months before
France in this multicenter, open–labeled RCT. After a run-in period of inclusion, and three in the NIAT group had chemotherapy completed
6 months, during which time patients were treated with the maximal 8 months, 2.5 years, and 6 years before inclusion. After 12 months, we
tolerated dose of NIAT (angiotensin–converting enzyme inhibitors deleted the time limit for the kidney biopsy, and we decreased the
and/or angiotensin 2 receptor blockers, diuretics, and statin), patients eGFR threshold down to 30 ml/min per 1.73 m2 to improve recruit-
were randomly assigned to 6-month therapy with NIAT plus rituximab ment. Sixty-nine patients had a renal biopsy taken ,2 years before
or NIAT alone (Figure 3). The goal of the NIAT group was to deter- inclusion. In the five remaining patients, the renal biopsies were taken
mine the percentage of nonimmunosuppressant-induced remissions, 25, 26, 28, 41, and 78 months before inclusion. Seven patients had
which was known to be high during the first 12 months.6 We delib- eGFR#45 and .30 ml/min per 1.73 m2. All patients gave written
erately opted for a short trial of 6 months to avoid any loss of a chance informed consent.
of patients receiving an immunosuppressive treatment in patients
who only received a supportive treatment. After the end of the ran- Procedures and Follow-Up
domized phase, patients were followed #24 months during a post– Patients received NIAT in association with 375 mg/m2 intravenous
RCT observational phase. The study was approved by an institutional rituximab on days 1 and 8 after randomization or NIATalone (Figure
review board in Paris, France (Comité de Protection des Personnes 3). We selected this dosing schedule on the basis of previous reports of
Ile-de-France XI). rituximab’s ability to induce proteinuria remission and CD19 deple-
tion.18,24 At the end of the 6-month randomized phase, referring
Patients physicians were free to reinfuse rituximab or shift immunosuppres-
Eligible patients were 18 years of age or older, had a biopsy-proven sant in patients of the NIAT-rituximab group or introduce an immu-
diagnosis established ,2 years before inclusion, had a urinary protein nosuppressant in patients of NIAT group, and patients were followed
excretion $3.5 g/d or a urinary protein-to-creatinine ratio $3500 #24 months during an observational period. The same antiproteinuric
mg/g, and had a serum albumin ,30 g/L for at least 6 months, despite drugs were allowed before and after randomization.
Study visits occurred at baseline, weeks 1 and 2,
and months 3 and 6 during the RCT. At each study
visit, clinical data and medications were recorded.
Blood and urine samples were collected at baseline
and months 3 and 6 for serum creatinine, serum
albumin, and proteinuria-to-creatinine ratio or
proteinuria excretion per day. PLA2R-Ab was
measured at baseline, day 8, and months 3 and 6.
CD19+ B lymphocyte counts were measured at
months 3 and 6 in the NIAT-rituximab group.
During the post–RCT observational phase,
visits occurred according to the habits of the
clinician in charge. Proteinuria, serum albumin,
serum creatinine, and immunosuppressive
treatment modifications were collected.
Figure 3. Study design. After a pre-inclusion period of 6 months during which NIAT
2 Data were collected in each of the 31 hospital
was optimized, patients were assigned either to NIAT plus Rituximab (375 mg/m , two
infusions at days 1 and 8) or to NIAT alone. Serum for anti-PLA2R-Ab determination nephrology units in a paper case report form and
was sampled at days 0 and 8, months 3 and 6, CD19 counts were determined at entered in a database located at Unité de Recher-
months 3 and 6; end points were assessed at month 6. The RCT was followed by an che Clinique-Centre de Recherche Clinique de
observational study during which follow-up was extended up to 24 months. IV, in- l’Est Parisien, an external and independent
travenous; R, rituximab. organization.

8 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org CLINICAL RESEARCH

Outcomes Sensitivity analyses were performed to check the effect of re-


The primary end point was the percentage of patients with complete or placement methods of missing values with missing data considered as
partial remission of nephrotic syndrome at 6 months of follow-up. (1) success (remission) in the NIAT group and failure (no remission)
Remission was defined according to 2012 KDIGO guidelines15 as (1) in the NIAT-rituximab group and (2) failure in the NIAT group and
complete in the case of urinary protein excretion ,500 mg/d or ,500 success in the NIAT-rituximab group. Additional analyses were per-
mg/g creatinine and (2) partial in the case of urinary protein excre- formed with missing data being replaced by last available data (pro-
tion ,3.5 g/d or ,3500 mg/g creatinine and $500 mg/g creatinine teinuria at 3 months) and available data under the hypothesis of data
with $50% reduction compared with baseline. Secondary end points missing completely at random.
were rate of proteinuria, serum albumin, serum creatinine, PLA2R- Additional logistic regression analyses were performed to identify
Ab levels, and SAEs. PLA2R-Ab was measured by using a quantitative potential prognostic factors of remission. The following variables of
ELISA (EuroImmune AG, Lubeck, Germany); THSD7A-Abs were interest were analyzed in univariate and multivariate analyses:
assessed by an immunofluorescence test (EuroImmune AG). Anti- treatment, age, sex, proteinuria, serum creatinine, serum albumin
body depletion was defined as complete disappearance of antibodies at baseline, and presence of PLA2R-Ab at baseline. In other explor-
in PLA2R-Ab–positive patients. Because albumin level may be an atory analyses, mean percentage changes from baseline of proteinuria,
earlier marker of response than end points defined by proteinuria serum albumin, and PLA2R-Ab levels at months 3 and 6 were plotted
only,23,27 we also considered a post hoc composite end point defined as and compared using a Wilcoxon matched pair signed rank test.
reduction of proteinuria .50% and increase of serum albumin level Statistical analyses were performed blinded to treatment allocation
.30% at month 6 of follow-up. on the basis of intention to treat and included all patients who received
Adverse events and unexpected changes in clinical or laboratory at least one dose of treatment and did not withdrawal consent. Safety
parameters were reported in patient case report forms and monitored population was defined as patients who received at least one dose of
up to complete resolution. All SAEs were monitored by Unité de treatment.
Recherche Clinique-Centre de Recherche Clinique de l’Est Parisien All tests were two sided, and P values ,0.05 were considered to
and reported to the sponsor. indicate statistical significance, except when Bonferroni correction
During the observational phase, remission defined according to was applied and mentioned. SAS V.9.3 software (SAS Institute Inc.,
the KDIGO and other variables were recorded before potential Cary, NC) was used for statistical analyses.
modification of treatment assigned at randomization (i.e., before any Findings from the trial are described in accordance with Consoli-
amendment of initial immunosuppressive treatment in the NIAT- dated Standards of Reporting Trials guidelines (www.consort-statement.
rituximab group or addition of an immunosuppressive treatment org). The trial was registered as the Evaluate Rituximab Treatment for
in the NIAT group). Follow-up was too short to record relapses. Idiopathic Membranous Nephropathy Study (Clinical Trials.gov no.
NCT01508468).
Statistical Analyses
On the basis of previous studies,17,18,25 rituximab was effective in
decreasing proteinuria as early as 3 months21,30 and achieving remis-
sion at 20 weeks17 to 1 year18,25 in 60%–80% of patients with PMN ACKNOWLEDGMENTS
and nephrotic syndrome persisting after 6 months of supportive ther-
apy. The trial was designed to establish whether rituximab was supe- We thank Nora Semai, Jennifer Piro, Julie Claudepierre, Benjamin
rior in terms of efficacy as assessed by the number of remissions. Laverdant, Matthieu Le lay, Anne Seguin, Dominique Damas, and
Assuming a remission rate of 20% in the NIAT group, the inclusion Elodie Drouet from the Clinical Research Department (Unité de
of 80 patients would provide 80% power at two-sided a of 0.05 to Recherche Clinique - Centre de Recherche Clinique de l’Est Parisien)
detect a 30% absolute increase in the remission rate (50%) under the at Saint Antoine University Hospital (Assistance Publique-Hôpitaux
assumption of 10% exclusion or dropout rates (Fisher exact test). de Paris, Université Pierre et Marie Curie) for trial monitoring and
Baseline characteristics of the study population were expressed as handling, preparation, and submission of all required research ethics
frequencies and percentages for qualitative variables and medians and and regulatory documents. We thank Emmanuel Roux for moni-
IQRs for continuous variables. Remission rates were expressed as toring and handling the biobank at Tenon Hospital (Prof. Isabelle
frequencies or percentages and their 95% CIs. All PLA2R-Ab titers not Brocheriou) and Christine Vial (UMR_S1155) for help in the prep-
achieving the 14-RU/ml detection threshold of the method were aration of the manuscript.
spiked at zero. PLA2R-Ab titer was considered as a continuous This study was funded by Programme Hospitalier de Recherche
variable, a binary variable (absence/presence), or at baseline only, a Clinique, French Ministry of Health grant AOM10089; European
categorical variable according to tertiles (,22.5 RU/ml, lowest; 22.5– Research Council ERC-2012-ADG_20120314 grant agreement
275.5 RU/ml, middle; $275.5 RU/ml, highest). Because at univariate 322947; Agence Nationale pour la Recherche Programme Blanc
analysis, tertiles 1 and 2 did not show any statistically significant SVSE1 (2012) Decision grant ANR-12-BSE1-0002-01; Fondation
difference, a binary variable was created (highest versus middle/lowest). pour la Recherche Médicale Equipe FRM 2012 grant; and 7th
Quantitative variables were compared by a t test or a Wilcoxon rank Framework Programme of the European Community contract 2012-
sum test, and categorical variables were compared by a Pearson chi- 305608 (European Consortium for High-Throughput Research in
squared test or a Fisher exact test. Rare Kidney Diseases). The sponsor of this study was Assistance

J Am Soc Nephrol 28: ccc–ccc, 2016 Rituximab for Membranous Nephropathy 9


CLINICAL RESEARCH www.jasn.org

Publique-Hôpitaux de Paris (Département de la Recherche Clinique Grassi C, Farina M, Bellazzi R: A randomized study comparing methyl-
et du Développement, Clinical Research and Development De- prednisolone plus chlorambucil versus methylprednisolone plus cy-
clophosphamide in idiopathic membranous nephropathy. J Am Soc
partment). Rituximab was given by Hoffmann-La Roche.
Nephrol 9: 444–450, 1998
This work was presented, in part, at the meeting of the American 9. Jha V, Ganguli A, Saha TK, Kohli HS, Sud K, Gupta KL, Joshi K, Sakhuja
Society of Nephrology in San Diego, California (November 3–8, 2015). V: A randomized, controlled trial of steroids and cyclophosphamide in
None of the funding sources played a role in the writing of the adults with nephrotic syndrome caused by idiopathic membranous
manuscript or the decision to submit it for publication. nephropathy. J Am Soc Nephrol 18: 1899–1904, 2007
Members of the Evaluate Rituximab Treatment for Idiopathic 10. Howman A, Chapman TL, Langdon MM, Ferguson C, Adu D, Feehally
J, Gaskin GJ, Jayne DR, O’Donoghue D, Boulton-Jones M, Mathieson
Membranous Nephropathy Research Group are listed in Supple-
PW: Immunosuppression for progressive membranous nephropathy: A
mental Material. UK randomised controlled trial. Lancet 381: 744–751, 2013
11. Chen Y, Schieppati A, Cai G, Chen X, Zamora J, Giuliano GA, Braun N,
Perna A: Immunosuppression for membranous nephropathy: A sys-
DISCLOSURES tematic review and meta-analysis of 36 clinical trials. Clin J Am Soc
K.D. has received grants from the French Ministry of Health and Roche (Basel, Nephrol 8: 787–796, 2013
12. Cattran DC, Greenwood C, Ritchie S, Bernstein K, Churchill DN, Clark
Switzerland). E.P. is a paid adviser for and has received lecture fees from Gilead
WF, Morrin PA, Lavoie S; Canadian Glomerulonephritis Study Group: A
Sciences (Foster City, CA); she has received research grants from Amgen, Inc.
controlled trial of cyclosporine in patients with progressive membra-
(Thousand Oaks, CA). T.S. is a paid adviser for and has received lecture fees
nous nephropathy. Kidney Int 47: 1130–1135, 1995
from AstraZeneca Pharmaceuticals (Wilmington, DE), Astellas, Bayer HealthCare 13. Cattran DC, Appel GB, Hebert LA, Hunsicker LG, Pohl MA, Hoy WE,
(Whippany, NJ), BMS, Lilly, MSD, and Sanofi US (Bridgewater, NJ); she has Maxwell DR, Kunis CL; North America Nephrotic Syndrome Study
received research grants from AstraZeneca Pharmaceuticals, Daiichi-Sankyo, Group: Cyclosporine in patients with steroid-resistant membranous
Eli Lilly (Indianapolis, IN), GlaxoSmithKline (Brentford, United Kingdom), nephropathy: A randomized trial. Kidney Int 59: 1484–1490, 2001
MSD, Novartis (Basel, Switzerland), and Sanofi (Bridgewater, NJ). P.R. is a 14. Praga M, Barrio V, Juárez GF, Luño J; Grupo Español de Estudio de la
paid adviser for Otsuka Pharmaceuticals France SAS (Rueil-Malmaison, Nefropatía Membranosa: Tacrolimus monotherapy in membranous
France), Bristol-Myers Squibb Company (Princetown, NJ), Amicus Thera- nephropathy: A randomized controlled trial. Kidney Int 71: 924–930,
peutics France (Paris, France), and Air Liquide (Paris, France); has received 2007
lecture fees from Shire (Belgium), Servier, Chugai Pharmaceutical Co. (Tokyo, 15. Kidney Disease: Improving Global Outcomes (KDIGO) Glomerulone-
phritis Work Group: KDIGO clinical practice guideline for glomerulo-
Japan), and the Mayo Clinic; and has received research grants from Amgen SAS
nephritis. Kidney Int Suppl 2: 186–197, 2012
(Boulogne-Billancourt, France) and Otsuka Pharmaceuticals France SAS.
16. van den Brand JA, van Dijk PR, Hofstra JM, Wetzels JF: Long-term
Hoffman-La Roche (Basel, Switzerland) provided rituximab for the study.
outcomes in idiopathic membranous nephropathy using a restrictive
treatment strategy. J Am Soc Nephrol 25: 150–158, 2014
17. Remuzzi G, Chiurchiu C, Abbate M, Brusegan V, Bontempelli M,
REFERENCES Ruggenenti P: Rituximab for idiopathic membranous nephropathy.
Lancet 360: 923–924, 2002
18. Fervenza FC, Cosio FG, Erickson SB, Specks U, Herzenberg AM, Dillon
1. Ponticelli C, Glassock RJ: Glomerular diseases: Membranous
JJ, Leung N, Cohen IM, Wochos DN, Bergstralh E, Hladunewich M,
nephropathy–a modern view. Clin J Am Soc Nephrol 9: 609–616, 2014
2. Beck LH Jr., Bonegio RG, Lambeau G, Beck DM, Powell DW, Cummins Cattran DC: Rituximab treatment of idiopathic membranous ne-
TD, Klein JB, Salant DJ: M-type phospholipase A2 receptor as target phropathy. Kidney Int 73: 117–125, 2008
antigen in idiopathic membranous nephropathy. N Engl J Med 361: 19. Bomback AS, Derebail VK, McGregor JG, Kshirsagar AV, Falk RJ,
11–21, 2009 Nachman PH: Rituximab therapy for membranous nephropathy: A
3. Hofstra JM, Fervenza FC, Wetzels JF: Treatment of idiopathic mem- systematic review. Clin J Am Soc Nephrol 4: 734–744, 2009
branous nephropathy. Nat Rev Nephrol 9: 443–458, 2013 20. Fervenza FC, Abraham RS, Erickson SB, Irazabal MV, Eirin A, Specks U,
4. Cravedi P, Remuzzi G, Ruggenenti P: Rituximab in primary membranous Nachman PH, Bergstralh EJ, Leung N, Cosio FG, Hogan MC, Dillon JJ,
nephropathy: First-line therapy, why not? Nephron Clin Pract 128: 261– Hickson LJ, Li X, Cattran DC; Mayo Nephrology Collaborative Group:
269, 2014 Rituximab therapy in idiopathic membranous nephropathy: A 2-year
5. Schieppati A, Mosconi L, Perna A, Mecca G, Bertani T, Garattini S, study. Clin J Am Soc Nephrol 5: 2188–2198, 2010
Remuzzi G: Prognosis of untreated patients with idiopathic membra- 21. Ruggenenti P, Cravedi P, Chianca A, Perna A, Ruggiero B, Gaspari F,
nous nephropathy. N Engl J Med 329: 85–89, 1993 Rambaldi A, Marasà M, Remuzzi G: Rituximab in idiopathic membra-
6. Polanco N, Gutiérrez E, Covarsí A, Ariza F, Carreño A, Vigil A, Baltar J, nous nephropathy. J Am Soc Nephrol 23: 1416–1425, 2012
Fernández-Fresnedo G, Martín C, Pons S, Lorenzo D, Bernis C, 22. Beck LH Jr., Fervenza FC, Beck DM, Bonegio RG, Malik FA, Erickson SB,
Arrizabalaga P, Fernández-Juárez G, Barrio V, Sierra M, Castellanos I, Cosio FG, Cattran DC, Salant DJ: Rituximab-induced depletion of anti-
Espinosa M, Rivera F, Oliet A, Fernández-Vega F, Praga M; Grupo de PLA2R autoantibodies predicts response in membranous nephropathy.
Estudio de las Enfermedades Glomerulares de la Sociedad Española de J Am Soc Nephrol 22: 1543–1550, 2011
Nefrología: Spontaneous remission of nephrotic syndrome in idiopathic 23. Ruggenenti P, Debiec H, Ruggiero B, Chianca A, Pellé T, Gaspari F,
membranous nephropathy. J Am Soc Nephrol 21: 697–704, 2010 Suardi F, Gagliardini E, Orisio S, Benigni A, Ronco P, Remuzzi G: Anti-
7. Ponticelli C, Zucchelli P, Passerini P, Cagnoli L, Cesana B, Pozzi C, phospholipase A2 receptor antibody titer predicts post-rituximab
Pasquali S, Imbasciati E, Grassi C, Redaelli B, Sasdelli M, Locatelli F: outcome of membranous nephropathy. J Am Soc Nephrol 26: 2545–
A randomized trial of methylprednisolone and chlorambucil in idio- 2558, 2015
pathic membranous nephropathy. N Engl J Med 320: 8–13, 1989 24. Cravedi P, Ruggenenti P, Sghirlanzoni MC, Remuzzi G: Titrating
8. Ponticelli C, Altieri P, Scolari F, Passerini P, Roccatello D, Cesana B, rituximab to circulating B cells to optimize lymphocytolytic therapy in
Melis P, Valzorio B, Sasdelli M, Pasquali S, Pozzi C, Piccoli G, Lupo A, idiopathic membranous nephropathy. Clin J Am Soc Nephrol 2: 932–
Segagni S, Antonucci F, Dugo M, Minari M, Scalia A, Pedrini L, Pisano G, 937, 2007

10 Journal of the American Society of Nephrology J Am Soc Nephrol 28: ccc–ccc, 2016
www.jasn.org CLINICAL RESEARCH

25. Ruggenenti P, Chiurchiu C, Brusegan V, Abbate M, Perna A, Filippi 29. Tomas NM, Beck LH Jr., Meyer-Schwesinger C, Seitz-Polski B, Ma H,
C, Remuzzi G: Rituximab in idiopathic membranous nephropathy: A Zahner G, Dolla G, Hoxha E, Helmchen U, Dabert-Gay AS, Debayle D,
one-year prospective study. J Am Soc Nephrol 14: 1851–1857, Merchant M, Klein J, Salant DJ, Stahl RA, Lambeau G: Thrombospondin
2003 type-1 domain-containing 7A in idiopathic membranous nephropathy.
26. Wagner MC, Campos-Bilderback SB, Chowdhury M, Flores B, Lai X, N Engl J Med 371: 2277–2287, 2014
Myslinski J, Pandit S, Sandoval RM, Wean SE, Wei Y, Satlin LM, 30. Hoxha E, Harendza S, Zahner G, Panzer U, Steinmetz O, Fechner K,
Helmchen U, Stahl RA: An immunofluorescence test for phospholi-
Wiggins RC, Witzmann FA, Molitoris BA: Proximal tubules have the
pase-A₂-receptor antibodies and its clinical usefulness in patients with
capacity to regulate uptake of albumin. J Am Soc Nephrol 27: 482–
membranous glomerulonephritis. Nephrol Dial Transplant 26: 2526–
494, 2016
2532, 2011
27. Ruggenenti P, Mise N, Pisoni R, Arnoldi F, Pezzotta A, Perna A,
31. Debiec H, Ronco P: PLA2R autoantibodies and PLA2R glomerular de-
Cattaneo D, Remuzzi G: Diverse effects of increasing lisinopril doses posits in membranous nephropathy. N Engl J Med 364: 689–690, 2011
on lipid abnormalities in chronic nephropathies. Circulation 107: 586–
592, 2003
28. Du Y, Li J, He F, Lv Y, Liu W, Wu P, Huang J, Wei S, Gao H: The diagnosis
accuracy of PLA2R-AB in the diagnosis of idiopathic membranous This article contains supplemental material online at http://jasn.asnjournals.
nephropathy: A meta-analysis. PLoS One 9: e104936, 2014 org/lookup/suppl/doi:10.1681/ASN.2016040449/-/DCSupplemental.

J Am Soc Nephrol 28: ccc–ccc, 2016 Rituximab for Membranous Nephropathy 11

Vous aimerez peut-être aussi