Vous êtes sur la page 1sur 13

Pediatric Nephrology

https://doi.org/10.1007/s00467-018-3970-y

EDUCATIONAL REVIEW

Mycophenolate mofetil for sustained remission in nephrotic syndrome


Uwe Querfeld 1 & Lutz T. Weber 2

Received: 23 November 2017 / Revised: 13 April 2018 / Accepted: 17 April 2018


# IPNA 2018

Abstract
The clinical application of mycophenolate mofetil (MMF) has significantly widened beyond the prophylaxis of acute and chronic
rejections in solid organ transplantation. MMF has been recognized as an excellent treatment option in many immunologic
glomerulopathies. For children with frequently relapsing nephrotic syndrome (FRNS) or steroid-dependent nephrotic syndrome
(SDNS) experiencing steroid toxicity, MMF has been recommended as a steroid-sparing drug. Uncontrolled studies in patients
with FRNS and SDSN have shown that many patients can achieve sustained remission of proteinuria with MMF monotherapy.
Three randomized controlled trials have similarly demonstrated that MMF is beneficial in these patients, but less effective than
the calcineurin inhibitors cyclosporin A or tacrolimus. Some, but not all, patients with steroid-resistant nephrotic syndrome
(SRNS) may also respond to MMF, usually given in combination with other drugs, with partial or complete remission. There are
important limitations to the interpretation and comparability of these studies including study design, sample size, patient selec-
tion, clinical endpoints, carry-over effects, and duration of follow-up. In all studies, MMF had relatively few side effects, no
nephrotoxicity, or no systemic toxicity. MMF is teratogenic, and contraceptive advice is required in females. There is a poor
correlation between MMF dose and mycophenolic acid (MPA) exposure and significant inter- and intra-patient variability in drug
pharmacokinetics. A higher estimated MPA-AUC0–12 target range than recommended for pediatric renal transplant recipients is
essential to prevent relapses. Therefore, therapy should be guided by drug monitoring to avoid relapses. Further studies are
needed to test the efficacy of MMF in inducing remission and, as part of a combination therapy, achieving sustained remission in
patients with SRNS.

Keywords Idiopathic nephrotic syndrome . Corticosteroids . Mycophenolate mofetil . Mycophenolic acid . Cyclosporin A .
Tacrolimus

Introduction accompanied by severe adverse effects, including, but not


limited to, the development of obesity, diabetes, arterial hy-
Idiopathic nephrotic syndrome (INS) is the most frequent glo- pertension, bone demineralization, aseptic bone necrosis, di-
merular disease in children. About 85% of affected patients minished growth, cataracts, infectious complications, and
have minimal change nephrotic syndrome (MCNS), which is thrombotic complications.
most often sensitive to treatment with corticosteroids (steroid- Therefore, non-steroid immunosuppressive agents have
sensitive nephrotic syndrome (SSNS)), with relapses occur- been empirically used for treatment of children with FRNS,
ring in 80–90% [1]. About 50% of patients with relapses have SDNS, and SRNS (recently reviewed by the Cochrane group)
frequently relapsing nephrotic syndrome (FRNS), and of [2] and their use has been recommended for patients
these, 30–50% steroid-dependent nephrotic syndrome experiencing steroid-related adverse effects by the expert
(SDNS). Prolonged use of corticosteroids is often KDIGO work group [3] and by an expert group of the
German Society for Pediatric Nephrology (GPN) [4]. The
published data of clinical trials of steroid-sparing agents indi-
* Uwe Querfeld cate that alkylating agents (cyclophosphamide, chlorambucil),
Uwe.querfeld@charite.de calcineurin inhibitors (cyclosporine, tacrolimus), levamisole,
rituximab, and mycophenolate mofetil induce or preserve re-
1
Pediatric Nephrology, Charité Campus Virchow, mission in FRNS and SDNS and might be considered as
13353 Berlin, Germany steroid-sparing agents, whereas azathioprine and mizoribine
2
University Children’s Hospital, Cologne, Germany have no significant therapeutic effect. Unfortunately,
Pediatr Nephrol

therapeutic use of all of these agents has been associated with Potential toxicities
distinct side effects, and there is currently no consensus on the
choice of a first-line steroid-sparing agent for patients with MMF has low systemic toxicity, and only few side effects
FRNS or SSNS [5]. have been reported in nephrotic patients. However, in combi-
We here review the experience with MMF in children with nation with other immunosuppressants, MMF may have more
the idiopathic nephrotic syndrome. serious side effects, especially infections. In transplanted pa-
tients, multifocal leukoencephalopathy has been linked to
MMF therapy [16, 17]. Of note, MMF has teratogenic poten-
Mycophenolate mofetil tial and in utero exposure has been associated with a specific
embryopathy; women should be counseled on the risk of this
Immune actions drug, and for women considering pregnancy, it is recommend-
ed to stop treatment with MMF at least 6 weeks before be-
Mycophenolate mofetil (MMF) is an ester prodrug of the im- coming pregnant [18].
munosuppressant mycophenolic acid (MPA) and is widely
used for maintenance immunosuppressive therapy in pediatric
renal transplant recipients. MPA acts as a potent, reversible, Principal limitations of clinical trials in patient
uncompetitive inhibitor of inosine monophosphate dehydro- with relapsing NS
genase (IMPDH), the key enzyme in the de novo purine bio-
synthesis in proliferating T and B lymphocytes, thereby sup- There are several important limitations which need to be con-
pressing cell-mediated immune responses and antibody for- sidered in the interpretation of results of clinical trials.
mation. In addition, MPA inhibits adhesion molecule glyco-
sylation and expression as well as lymphocyte and monocyte & The INS may follow its natural course and resolve with
recruitment [6]. MPA also induces apoptosis of immune cells. time. Treatment effects may thus be falsely attributed to a
Proliferating T and B cells are preferentially dependent on particular medication.
the de novo purine synthesis. This is why MPA has a quite & The disease affects mainly boys, and results of all studies
specific effect on proliferating lymphocytes. In contrast, other may be biased by a preponderance of male participants;
cells (e.g., brain cells) exclusively utilize the so-called salvage drug effects could be different in girls.
pathway that is based on recycling of purine bases [7]. In the & The response to steroids may be variable, even within
majority of eukaryotic cells, purine synthesis can be achieved histological categories on renal biopsy. Thus, and for rea-
by both pathways. sons unknown, patients with MCNS may be steroid resis-
tant or become steroid resistant, and patients with FSGS
Non-immune actions may be steroid responsive and may become steroid resis-
tant. It is unknown to what extent patients with primary or
The clinical application of MMF has significantly widened late steroid resistance, even with MCNS, will respond to
beyond the prophylaxis of acute and chronic rejections in solid other immunosuppressive drugs. Thus, inclusion of these
organ transplantation. For example, it has become part of the patients in clinical studies may be problematic.
first-line therapy in lupus nephritis [8, 9] and meanwhile has & All steroid-sparing drugs used for treatment of the NS
been recognized as an excellent treatment option in many have strong immunosuppressive properties. Very little is
immunologic glomerulopathies, firstly, because MPA pos- known about interactions of these immunosuppressants
sesses the immunosuppressive properties described above. when given in combination or in sequence [19].
Moreover, MPA has a remarkable effect also on non- However, drug effects may last for unknown periods of
immune cells and counteracts the proliferation of mesangial time, by far exceeding the time of medication intake.
cells, the expansion of mesangial matrix, and the foot process Thus, a single dose of rituximab may lead to a significant
effacement of podocytes [10–14]. depletion of memory B cell subsets measured after 1 year
Similar to lymphocytes, mesangial cells are partially de- [20]. Typically, patients with unremitting FRNS or SDNS
pendent on de novo purine biosynthesis and thus susceptible receive multiple courses of different steroid-sparing agents
for MPA treatment. Additionally, MPA can inhibit apoptosis within a few years; if such patients are enrolled into clin-
in podocytes and seems to be beneficial in preserving the ical studies, their drug history may be a major factor de-
expression of nephrin and podocin; by attenuation of uroki- termining response to treatment. As an example, in the
nase receptor expression, MPA leads to decreased foot process crossover trial conducted by the GPN, the efficacy of
effacement [15]. Thereby, MPA seems to exert a direct MMF was significantly different when the patients re-
antiproteinuric effect that makes it a valuable drug in the treat- ceived the drug in the first year of the study or in the
ment of nephrotic syndrome. second year after switching from cyclosporin A (CsA) to
Pediatr Nephrol

MMF. There were significantly more relapses per patient those with a low exposure (p = 0.03) and not different com-
per year with MMF compared to CsA therapy during the pared to patients treated with CsA (p = 0.32).
first year (1.10 vs. 0.24; p = 0.034), but not during the Sinha and colleagues performed an RCT comparing MMF
second year (0.40 vs. 0.20; p = 0.14). It is currently un- treatment with tacrolimus (TAC) in children with steroid-
known to what extent and how long such carry-over ef- resistant nephrotic syndrome (early or late steroid resistance
fects may affect treatment results. with minimal changes or focal segmental sclerosis on biopsy)
& Finally, drug pharmacokinetics may show large inter- and achieving remission for 6 months with Tac, prednisolone, ac-
intra-individual variability and study results may differ, companied by enalapril and calcium carbonate medication
depending on the use of therapeutic drug monitoring [23]. During remission, patients were randomized to receive
(TDM). Unfortunately, TDM has not been standardized MMF or continue with Tac for 12 months. The proportion of
and different algorithms are used to estimate MPA-AUC. patients with a favorable outcome (sustained remission, infre-
Preferably, blood samples should be analyzed within quent relapses) at 1 year was significantly lower (44.8%) in
hours and results reported on the same day. Differences the MMF group than in the Tac group (90.3%). The authors
in local TDM methodology and logistics may therefore concluded that replacing tacrolimus with MMF after 6 months
impact results of clinical studies, especially in multicenter of tacrolimus therapy for SRNS is associated with significant
trials. risk of frequent relapses or recurrence of resistance.
In these controlled studies, MMF was generally well toler-
ated. All RCTs reported mostly minor side effects, although
there was a higher tendency for serious adverse effects in the
Randomized clinical trials MMF group in the study by Sinha et al. Interestingly, hemo-
globin levels were significantly lower in patients treated with
There have been only a few randomized clinical trials (RCTs) CSA compared to MMF in both CsA studies, and there was a
in children with either frequently relapsing or steroid- higher incidence of anemia in patients treated with Tac than
dependent SSNS (Table 1). Dorresteijn et al. enrolled 24 chil- with MMF. Altogether, the calcineurin inhibitors CsA and Tac
dren for 12 months and observed a higher relapse rate in performed better than MMF in all randomized trials.
patients treated with MMF than with CsA (p = 0.08) after
12 months of treatment [21]. While this study was underpow-
ered as acknowledged by the authors, the RCT conducted by Uncontrolled studies evaluating MMF
the German Society for Pediatric Nephrology, which enrolled monotherapy in FRNS or SDNS
60 patients, confirmed the higher efficacy of CsA [22]. In this
crossover study, with an adequate sample size based on a non- In these hospital-based cohort studies (Table 2), MMF was
inferiority power calculation, every patient received either introduced as a steroid-sparing agent. In most studies, patients
MMF or CsA for 12 months, followed by treatment with the had been previously treated with other immunosuppressants.
other drug for a second year. No relapses occurred in 85% of The clinical endpoints in these studies were different, making
patients during CsA therapy and in 64% of patients during them hardly comparable. Further issues limiting the relevance
MMF therapy (p = 0.06). Probably due to carry-over effects of these data for clinical decision making are the small number
of the more potent medication with CsA, more relapses per of subjects involved, lack of randomization, and only a short
patient per year occurred with MMF than with CsA during the follow-up period in most studies. However, MMF was bene-
first year (p = 0.03), but not during the second year (p = 0.14). ficial in all studies, associated with a higher remission/lower
Similarly, the time without relapse was significantly longer relapse rate or lower steroid requirements if compared to a
with CsA than with MMF during the first year (p < 0.05), previous treatment period with corticosteroids or in patients
but not during the second year (p = 0.36). switched from CsA. MMF was equally [33] or less [35] effi-
Importantly, treatment with both medications was adjusted cacious compared to Tac treatment in two uncontrolled trials.
by pharmacokinetic monitoring every 6 months. The MMF Only few MMF-related side effects were reported in all of
starting dose was 1000 mg/m2/day in two doses and the target these studies.
plasma MPA trough level 1.5–2.5 μg/ml, while CsA was giv-
en at a dose of 150 mg/m2 in two doses with a target trough
level of 80–100 ng/ml. A post hoc analysis revealed that pa- Uncontrolled studies including SRNS patients
tients with low mycophenolic acid exposure (AUC < 50 μg·h/ evaluating MMF monotherapy
ml) experienced 1.4 relapses per year compared with 0.27
relapses per year in those with high exposure (AUC > Several pediatric centers have published their experience with
50 μg·h/ml; p < 0.05). The relapse-free time was significantly MMF treatment in cohorts including patients with SSNS and
higher in patients with a high MMF exposure compared to SRNS (Table 3).
Pediatr Nephrol

Table 1 MMF treatment of nephrotic syndrome in childhood: randomized clinical trials

Author/year/country Comparator Primary endpoint Type of study N (age) Dose per day Outcome
[ref.] (clinical Histological
classification) classification

Dorresteijn MMF or CsA for Number of Prospective 24 MMF 1200 mg/m2 Relapse rate in the
et al./2008/Holla- 12 months relapses open label (3.7–- (n = 12) or CsA MMF group
nd [21] (8 FRNS, 17.5 years) 4–5 mg/kg (n = 12) 0.83/year vs.
16 SDNS) All patients for 12 months 0.08/year in the CsA
SSNS, group (p = 0.08)
MCNS
Gellermann MMF vs. CsA, each Number of Prospective, 60 (3.3–17.0 y)MMF 1000 mg/m2 No relapses in 85% of
et al./2013/Germ- drug for 12 months, relapses open label, All patients starting dose; target patients during CsA
any [22] then crossover 24 month- SSNS, plasma MPA trough therapy and in 64%
s, MCNS level 1.5–2.5 μg/ml) of patients during
crossover or CsA 150 mg/m2 MMF therapy
at in 2 doses (target (p = 0.06). More
12 months trough level relapses per patient
(14 FRNS, 80–100 ng/ml) per year with MMF
46 SDNS) than with CsA
during the first year
(p = 0.03), but not
during the second
year (p = 0.14)
Sinha MMF or Tac for Proportion of Prospective, 60 (2–6.6 years) Tac 0.1 to 0.15 mg/kg Better outcome with
et al./2017/India 12 months, patients with open label 56.7% or MMF tacrolimus (90.3%)
[23] maintaining complete or for MCNS, 750–1000 mg/m2 vs. MMF (44.8%)
remission of partial 12 months 43.3% FSGS Prednisolone (p = 0.0002)
proteinuria; SRNS in remission with (60 SRNS) tapered to a dose of
complete or partial or without (46.7% 0.2 to 0.3 mg/kg
remission after infrequent had initial q.o.d. for 12 months
6 months of therapy steroid-- steroid in both groups.
with tacrolimus sensitive resistance)
relapses

CsA cyclosporine A, q.o.d. every other day, Tac, tacrolimus, y years, MMF mycophenolate mofetil, SDNS steroid-dependent nephrotic syndrome, SRNS
steroid-resistant nephrotic syndrome, FRNS frequently relapsing nephrotic syndrome, MCNS minimal change nephrotic syndrome

Studies describing MMF use in other glomerulopathies or free survival time shorter in patients treated with MMF after
with incomplete description of medication data or available 18 months of observation [47]. Similarly, in 24 children with
only in abstract form [44–46] have not been included in these SRNS who had received 2–4 doses of rituximab, treatment
tables. MMF was beneficial and had a steroid-sparing effect in with MMF in 15 patients resulted in complete remission in
all studies, but more effective in maintaining remission in 10 and partial remission in 5 patients within 12 months [35].
patients with SSNS. However, a varying percentage of pa- In 9 children with SDNS treated with rituximab, Ito et al.
tients with SRNS also achieved remission with MMF mono- found lower relapse rates in patiens receiving MMF as main-
therapy. Taken together, these studies (again highly varying in tenance therapy than in historic controls without maintenance
their design) suggest that MMF is a valuable pharmaceutical [48].
option in patients with SSNS and/or SRNS. Gellermann et al. reported long-term follow-up observa-
tions of 23 children with primary SRNS associated with
FSGS who achieved initial remission with MP pulse therapy
MMF in combination therapy/sequential and CsA. Maintenance therapy included CsA, ACE/ARB, and
therapy for SRNS tapering of corticosteroids. In 18 patients, mycophenolate mo-
fetil (MMF) (adjusted to achieve MPA trough concentrations
In a prospective study, Fujinaga et al. compared the efficacy of > 2 μg/ml) was sequentially added, and 16 patients were con-
MMF (adjusted to a pre-dose MPA level of 2–5 μg/ml) and verted to MMF monotherapy. During a follow-up time of 7
CsA (adjusted to a 2-h post-dose level of 400–500 ng/ml) in (1.7–16.5) years, sustained remission could be achieved in all
29 children with SDNS who had previously been maintained patients [49].
on CsA treatment (for > 4 years on average) and received one In a standardized protocol, Nibikakhsh et al. treated 37
dose of rituximab. The relapse rate was higher and the relapse- patients with primary SRNS with cyclosporine A (CyA) and
Pediatr Nephrol

Table 2 MMF treatment of steroid-sensitive nephrotic syndrome in childhood: uncontrolled studies

Author/year/ Comparator Primary Type of study N (age) Dose (per day) Main outcome
country [reference [previous endpoint (clinical Histological
number] treatment with classification) classification
steroid-sparing
agents]

Bagga MMF for Relapse rate Prospective 19 29 mg/kg/day, Decreased mean relapse rate (from
et al./2003/India 12 months single (2.6–- 12 months 6.6 to 2 episodes/year), but
[24] [CyP, LEV] center 11.2 years) treatment failure in 3 patients
(SDNS) 10 MCNS
3 FSGS
6 unknown
histology
Novak MMF for Relapse rate Retrospective 21 (2–17 years) 1200–2000 mg/m2 Reduction in relapse rate from
et al./2005/USA 1.0 ± compared single 0.80 ± 0.41 to 0.47 ± 0.43 relapses
[25] 0.5 years to steroids center per month (p < 0.02).
(0.2–- (SDNS)
2.0 years)
[None]
Hogg MMF for Remission Prospective 33 (2–15 years) 1200 mg/m2 24 patients in remission for 6 months
et al./2006/USA 6 months multicenter (75%)
[26] [Multiple] (27 FRNS; 6
SDNS)
Okada MMF for Remission Prospective 11 750 and 1000 mg/m2 Complete remission in 7, fewer
et al./2007/Japa- 12 months compared single (10–- relapses in 2, no effect in 1 patient
n [27] [CsA, to CsA center 22 years)
multiple] (conversion
from CsA)
(FRNS)
Fujinaga MMF for Relapse rate Prospective 12 (5–19 years) 610 ± 95 mg/m2/12 h, 9 patients (75%) could be weaned off
et al./2007/Japa- 12 months single adjusted to MPA CsA. The relapse rate decreased
n [28] [CsA, 7 MZR] center trough levels of from 2.7 ± 1.6 to 0.6 ± 0.9
(conversion 2.4 ± 1.1 μg/ml episodes/year. The probability of
from CsA) maintaining remission without
(SDNS) relapses on MMF therapy was
lower in patients whose average
trough MPA levels were less than
2 μg/ml (p < 0.05)
Afzal MMF for 14.3 Relapse rate Retrospective 42 26.5 mg/kg 50% or more reduction in relapse rate
et al./2007/India (6–45) after single (2.6–- in 76.2% and sustained remission
[29] months 6 months center 15.6 years) in 21.4% of patients.
[35 LEV, 37 compared (SDNS)
CyP] to past
6 months
Fujinaga MMF for 19 Relapse rate Retrospective 26 1000 mg/m2 adjusted 12-monthly relapse rates decreased
et al./2009/Japa- (7–42) single (5.8–- to trough levels of from 2.5 ± 1.4 to 0.8 ± 1.2
n [30] months center 20.5 years) > 2 μg/ml [19 episodes (p < 0.01). 15 patients
[CsA, 13 (SDNS) MCNS, 7 FSGS] weaned off CsA (58%). 12
MZR] patients had been included in
previous study [28]
Baudouin MMF for Relapse Prospective 24 600 mg/m2 for 7 days, 4 patients relapsed during the first
et al./2012/Fran- 12 months during the single (2.8–- then 1200 mg/m2 6 months and 2 at months 8 and
ce [31] [Alkylating first center 14.4 years) 11; 19 patients free of relapse
agent, 17 6 months (SDNS) during the first 6 months
LEV] of MMF
treatment
Banerjee MMF for Change in Retrospective 46 (6.57 (± 3.0) 20–30 mg/kg After 1 year of, 32 (70%) patients
et al./2013/India 12 months steroid multicenter years) had reduced steroid requirement
[32] [CyP, LEV, 4 require- (SDNS) and 20 of them were able to stop
CNI] ment steroids
Wang MMF or Tac Remission Prospective 65 MMF 20–30 mg/kg No difference in remission rates
et al./2016/Chi- for rate, single (5.3–- during the first and second
na [33] 12 months relapse center, not 6.0 years) 6-month follow-ups between the
Pediatr Nephrol

Table 2 (continued)

Author/year/ Comparator Primary Type of study N (age) Dose (per day) Main outcome
country [reference [previous endpoint (clinical Histological
number] treatment classification
with steroid- classifica-
sparing tion)
agents]

[None] rate random- 30 MMF, 35 Tac Tac 0.05–0.15 mg/kg TAC and MMF groups (97.1 vs.
compared ized adjusted to trough 90% at month 12) and in relapse
to Tac (FRNS or levels of 5–10 μg/l rates during follow-up
SDNS)
Dehoux MMF Relapse rate Single center 96 600 mg/m2 for 7 days, 67% reduction in relapse rate, 40%
et al./2016/Fran- (minimum Compared retrospec- (4.2–- then 1200 mg/m2 reduction of the cumulative annual
ce (Dehoux treatment to previous tive 10.4 years) dose of prednisone after
et al., 2016) time steroid 1995–2012 12 months
12 months) treatment (SDNS)
[Multiple]
Jellouli MMF Relapse rate Single center 30 MMF 1200 mg/m2 The average rate of relapse 1.75/year
et al./2016/Tuni- [Multiple] compared retrospec- (3.5–- before MMF and 0.45/year during
sia [34] to previous tive 16 years) MMF therapy (p < 0.0001).
steroid (SDNS) Responding patients were younger
treatment at the onset of MMF (8.57 versus
12.83, p = 0.009)
Basu MMF or Tac or Relapse rate Single center 340 MMF 1200 mg/m2 Relapse-free survival significantly
et al./2016/India LEV For and retrospec- (1.5–- Tac 0.1–0.2 mg/kg longer with Tac compared to
[35] 18 months relapse-- tive single 13 years) adjusted to trough MMF (61.7 versus 38.5%;
[None] free center levels of 5–7 ng/ml p < 0.001) or LEV (61.7 versus
survival (FRNS, Levamisol 2.5 mg/kg 24%; p < 0.0001)
with each SDNS) (q.o.d.)
drug

CNI calcineurin inhibitor, CsA cyclosporine A, LEV levamisole, Tac tacrolimus, MZR mizoribine, CyP cyclophosphamide, SDNS steroid-dependent
nephrotic syndrome, FRNS frequently relapsing nephrotic syndrome, MMF mycophenolate mofetil

alternate day prednisolone for 6 months, resulting in complete Tac, and MMF with a 75% response rate [53]. Some other
remission in 12 and partial remission in 2 patients. The other studies could not be evaluated since data were only available
23 cases were then treated with CyA (5 mg/kg/day) and MMF in abstract form.
(30 mg/kg/day) for 6 months. Complete remission was ob- Taken together, these studies indicate that MMF is also
served in 11 cases (47.82%) and partial remission in 2 cases useful as part of a combination/sequential therapy in select-
(8.7%) [50]. ed patients, especially in those with secondary steroid re-
Hibino et al. reviewed the 3-year outcome of 91 patients sistance or with primary steroid resistance achieving remis-
with SDNS or SRNS treated with a standard protocol, which sion. Studies reporting long-term observations [49, 51, 53]
added MMF (40 mg/kg/day) to maintenance therapy with found long-term remission rates varying between 67 and
CsA and prednisolone and/or MP pulse therapy in patients 100%. These differences could be related to differences in
with continuing relapses. In patients with SRNS, they could patient selection or other factors including dosing, compli-
achieve remission rates of 100% (6/6) in MCNS and 67% ance, genetic differences in patient cohorts, and histological
(8/12) in FSGS [51]. diagnosis. However, MMF seems to be a valuable treatment
In a prospective study including children with steroid- option for this difficult-to-treat group of patients, typically
and Tac-resistant (n = 10) or TacTAC-sensitive but frequent- with FSGS, and more favorable results have been found
ly relapsing nephrotic syndrome (n = 8), Wu et al. added with longer observation periods, suggesting a beneficial
either CyP (n = 6) or MMF (n = 5) or leflunomide (LEF; effect of long-term maintenance with MMF in patients tol-
n = 7) as a third immunosuppessant in a randomized manner. erating this medication. Here, MMF has a place in combi-
This triple therapy was effective in achieving short-term nation with ACE/ARB, calcineurin inhibitors, and pulse
remission and preventing long-term relapses of proteinuria therapy with methylprednisolone to achieve remission or
during 1-year follow-up, without a significant difference minimize proteinuria; an aggressive approach seems justi-
between groups [52]. Kim et al. treated 8 children with fied to prevent or at least slow down progressive loss of
SRNS with a sequential therapy including corticosteroids, GFR.
Pediatr Nephrol

Table 3 MMF treatment of steroid-sensitive and steroid-resistant nephrotic syndrome: uncontrolled studies

Author/year/country Comparator Primary Type of study N (age) Dose per day Main outcome
[reference] [previous endpoint (clinical Histological
treatment with classification) classification
steroid-sparing
agents]

Barletta et al./2003/USA MMF for Retrospective 14 (3.4–15.2 years) The mean number of relapses
[36] 12 months Relapses/- single center 5 MCNS 800–1200 m- was reduced from 2.85
[CsA (n = 10), year (9 SDNS 5 9 IgM-N g/m2 (± 0.4) prior to MMF to
CyP] SRNS) 1.07 (± 0.3) after
12 months of MMF
treatment (p < 0.01)
Montané MMF for Change in Retrospective 9 (2–15 years) 250–500 mg/m2 Up/c was 72% below
et al./2003/USA [37] 24 months proteinuria single center 9 FSGS baseline (p < 0.01) after
[MP, CyP, CsA, (SRNS; all 6 months; 3 patients had a
Tac] on ACE or complete remission by
ARB; 6 months, six had partial
pre-treatment remission
with MP)
Gellermann and Switch from Remission Retrospective 7 (9–16 yeras) Initially All patients (6 out of 7
Querfeld/2004/Germ- CsA to single center 6 MCNS 1000 mg/m2 children switched to MMF
any [38] MMF (6 SSNS, 1 1 FSGS adjusted to and 1 patient with SRNS
MMF for SRNS) trough MPA receiving MMF in addition
15–- levels of 1.5 to to CsA) achieved
39 months 4.5 mg/ml long-term remission
[CyP, LEV; MP,
plasmaphere-
sis in 1
patient with
FSGS]
Ulinski Switch from Remission Prospective 9 (3 to 16 years) Up to All SDNS patients stayed in
et al./2005/France [39] CsA to single center 1 g/1.73 m2 remission while 2 children
MMF (7 SDNS, 2 twice daily for with SRNS had no change
SRNS) 261 ± 183 day in level of proteinuria;
GFR increased from 76 ± 5
to 119 ± 6 ml/min per
1.73 m2
Mendizabal MMF for Remission Prospective 26 (5–17 years) 1200 mg/m2 Only one patient with SRNS
et al./2005/Spain [40] 6 months single center 11 MCNS adjusted to achieved complete
[MP, CsA, (21 SDNS, 5 13 FSGS trough MPA remission. In patients with
CyP] SRNS) 1 DMP levels of SDNS, steroid sparing was
1 MGN 2.5–5 μg/ml achieved in 15 and 9
remained in remission on
MMF monotherapy. In
patients with SDNS,
average trough MPA levels
< 2.5 μg/ml were
associated with double the
risk of relapses compared
to patients with levels
> 2.5 μg/ml
De Mello MMF for Complete or Retrospective 52 (2–17 years) 500–600 mg/m2 Complete remission were
et al./2010/Brazil [41] 6 months partial single center 30 FSGS achieved in 12 (23.1%),
[34 CsA] remission (SRNS) 19 MCNS partial remission 19
3 CGN (36.5%), while 21 (40.4%)
patients had no benefit
Li et al./2010/China [42] MMF for Remission Prospective 24 (8–23 months) 25–30 mg/kg Of these patients < 2 years of
6–12 months single center 15 DMP daily age, 8 had primary SRNS,
with (8 primary 4 IgM-N 16 late steroid resistance.
stepwise SRNS, 16 2 MMF treatment resulted in
reduction of late SRNS) proliferative-- complete remission in 15,
prednisone sclerotic partial remission in 6, no
3 FSGS response in 3
Pediatr Nephrol

Table 3 (continued)

Author/year/country Comparator Primary Type of study N (age) Dose per day Main outcome
[reference] [previous endpoint (clinical Histological
treatment classification
with steroid- classifica-
sparing tion)
agents]

Hassan et al./2013/UK MMF for Remission Retrospective 73 (7.9–13.6) 1200 mg/m2 MMF was effective in 45 of
[43] 2.1 years single center 73 (62%) patients
(0.8–- (73% SDNS, (complete remission in 36
3.2 years) 18% FRNS, (49%) for > 2 years).
2% SSNS, Partial remission (with
7% SRNS) complete steroid
withdrawal) in 2 (3%)
children for > 2 years.
Complete remission for 1
to 2 years in 7 (10%)
children

CsA cyclosporin A, CyP cyclophosphamide, DMP diffuse mesangial proliferation, GN chronic glomerulonephritis, LEV levamisole, MGN membranous
glomerulonephritis, IgM-N IgM nephropathy, MP methylprednisolone, prol-scerot proliferative and sclerotic lesions, y years, Up/c urine protein/
creatinine ratio, SSNS steroid-sensitive nephrotic syndrome, SDNS steroid-dependent nephrotic syndrome, MMF mycophenolate mofetil, FSGS focal
segmental glomerulosclerosis, FRNS frequently relapsing nephrotic syndrome

Side effects clinical trials to assess efficacy of MMF for patients in


these different diagnostic categories.
MMF was tolerated well by most patients included in the & Duration of therapy: It is currently unclear how long pa-
published studies. Side effects were highly variable and most- tients with FRNS, SDNS, or SRNS should be treated with
ly minor; only very few patients had to discontinue medica- MMF. While the therapeutic goal is to achieve sustained
tion. In the controlled trials (Table 1), the frequency of overall remission, defined as absence of proteinuria, there are cur-
side effects of MMF treatment was similar to that of treatment rently no biomarkers which are predictive of remission or
with CsA or tacrolimus, respectively. Diarrhea and abdominal relapse. Many studies have described rapid relapse of the
pain, frequently reported in children treated with MMF after nephrotic syndrome after discontinuation of MMF. Since
kidney transplantation [54], were infrequently observed. In prevention of a relapsing course is essential, treatment
the two largest uncontrolled single center retrospective stud- should therefore be planned for several years, especially
ies, the rate of gastrointestinal symptoms was 3.1% in the in patients with FSGS/SRNS.
study by Basu et al. (n =340) and 11.2% (6% only mild) in
the study of Dehoux et al. (n = 74) [16, 17]. In the study by
Hassan et al., which included SRNS patients, the rate of gas-
trointestinal side effects was 9% [18]. Pharmacokinetics

Pharmacokinetic therapeutic drug monitoring (TDM) in gen-


A critical view of published studies eral has the aim to optimize individual drug treatment. One
necessary precondition is an association of the drug’s pharma-
& Quality: In general, there are important limitations to the cokinetics and its clinical efficacy and/or toxicity. TDM is also
interpretation and comparability of published studies in- reasonable when there is high interindividual variability of
cluding study design (mostly uncontrolled), (small) sam- drug concentrations, when drug-drug interactions are present,
ple size, patient selection (non-randomized), clinical end- and when compliance needs to be monitored. All these argu-
points, carry-over effects, and duration of follow-up. ments apply to MMF [55].
& MMF efficacy: It is not possible to determine efficacy of There is a poor correlation between MMF dose and MPA
MMF from the published uncontrolled studies, which are exposure and significant inter- and intra-patient variability in
often further limited by a mix of patients with different drug pharmacokinetics [56]. This may be explained by genetic
diagnostic categories (SSNS, SDNS, FRNS), differences polymorphisms in key enzymes and drug transporters, involv-
in previous treatment regiments, and times of follow-up. ing uridine diphosphate glucuronosyltransferase enzymes, or-
There is clearly a need for more well-designed controlled ganic anion transport polypeptides, and multidrug resistance-
Pediatr Nephrol

associated protein 2. These genetic polymorphisms are asso- Bayesian estimators might be the tool of choice to perform
ciated with the metabolism, efficacy, and toxicity of MPA, TDM in this patient population.
thus resulting in different MPA exposures and patient out- In summary, TDM of MMF plays an important role in
comes [57]. For example, multidrug-resistant protein-2, children with nephrotic syndrome since several studies have
encoded by the MRP-2 gene, is an ATP-dependent efflux shown that a higher estimated MPA-AUC0–12 target range (>
transporter. The predominant role of MRP-2 is to export or- 45–50 mg × h/l) compared to the population of pediatric renal
ganic anions and xenobiotics out of cells and into the bile, transplant recipients (30–60 mg × h/l) is essential to prevent
urine, and intestinal lumen. This transporter is thus responsi- relapses. If not routinely performed, TDM should therefore be
ble for the biliary excretion of the major metabolite of MPA, considered for children experiencing relapses during treat-
the pharmacologically inactive 7-O-MPA glucuronide ment with MMF monotherapy for SSNS or with MMF in
(MPAG). Subsequent enterohepatic recirculation leads to sec- combination therapy for SRNS. As previously stated, high
ondary increases in plasma MPA concentrations 6–12 h post- intra-individual variability of the drug may result in
dose, which are estimated to contribute 40% (range 10–60%) underdosing, drug-drug interactions may be present when
to MPA exposure. Single nucleotide polymorphisms leading used in combination with other immunosuppressants, and
to altered MRP-2 activity may influence this process and compliance may be an issue; in these situations, TDM is an
therefore individually affect MPA exposure. extremely valuable tool for clinical decision making.
Importantly, exposure to MPA needs to be higher in chil-
dren with nephrotic syndrome to be efficacious than in pedi-
atric renal transplant recipients. In the latter, the usual target Is MMF useful in inducing remission?
MPA-AUC (area under the concentration versus time curve)
derived from 12-h pharmacokinetic profiles reflecting total So far MMF has been used for maintenance therapy, either in
body drug exposure is 30–60 mg × h/l [58]. In the group of patients with FSNS or in patients with SDNS in remission
children with nephrotic syndrome, however, target MPA- after a course of corticosteroids, or as a steroid-sparing agent
AUCs of > 45–50 mg × h/l were needed to preserve remission in patients with ongoing SDNS or SRNS achieving complete
[49, 59–61]. These studies also showed that higher MPA ex- or partial remission with other medication. It is unknown
posure was not associated with more side effects in children whether MMF alone or in combination is helpful in inducing
with INS. remission. The ongoing INTENT Study in Germany is a ran-
One obvious reason for this phenomenon doubtlessly is domized prospective trial to evaluate the potential steroid-
that immunosuppressive therapy after solid organ transplanta- sparing role of MMF in the initial treatment of steroid-
tion in general is a combined therapy whereas in FRNS or sensitive idiopathic nephrotic syndrome in children
SDNS, MMF often is used as monotherapy. This aspect is of (EudraCT No. 2014-001991-76). The INTENT Study trans-
some importance as far as the question of the performance of fers the beneficial effects of MMF in preserving remission to
TDM in the cohort of children with nephrotic syndrome is the period of initial treatment by substituting prednisone with
concerned. AUC monitoring is laborious and costly. The value MMF for the rest of the initial treatment period of 12 weeks
of trough-level monitoring is challenged by poor correlation (as soon as clinical remission is achieved). If this approach
with AUC in the transplant cohort [62], and interference with proves to be not inferior to the standard treatment with pred-
concomitant medication, especially cyclosporine A [63]. In nisone (in terms of time to first relapse and number of relapses
children with nephrotic syndrome, however, an association within 2 years of follow-up), it has the potential to change the
of MPA trough-level monitoring with efficacy has been dem- initial standard treatment of nephrotic syndrome to a less toxic
onstrated with higher trough levels resulting in higher rates of approach.
remission [28, 61]. Likewise, the use of limited sampling In conclusion, MMF is a valuable therapeutic option in
strategies to calculate AUC values has been validated to pre- children with SSNS exeriencing frequent relapses or steroid
dict clinical outcome [22, 61], thereby offering useful alterna- dependency. As a steroid-sparing agent, MMF is recommend-
tives to full-time pharmacokinetic monitoring. Limited sam- ed for patients with FRNS/SDNS experiencing steroid toxicity
pling strategies have also been used to develop Bayesian esti- by the KDIGO expert working group and the GPN expert
mators for TDM of MMF in children with nephrotic syndrome group. MMF has immunosuppressive and antiproteinuric ef-
[64, 65]. Bayesian estimators have the advantage of also tak- fects, is generally well tolerated, and lacks nephrotoxicity.
ing other factors—apart from pure pharmacokinetic parame- Therapy should be guided by drug monitoring to avoid re-
ters—into account when estimating drug exposure such as lapses due to underdosing. In SRNS patients, MMF is also
age, gender, renal function, and serum albumin concentra- beneficial in selected patients, but more experience is required
tions. Since INS is characterized by a relapsing course with to establish the most effective combination or sequential ther-
changing biomarkers with concomitant influence on MPA apy to achieve complete or partial remission. MMF is terato-
pharmacokinetics (e.g., serum albumin concentrations), genic and contraceptive advice is required in females. As in all
Pediatr Nephrol

patients requiring long-term immunosuppression, regular e) Exposure to MPA needs to be higher in children with
follow-up and a high index of suscpicion are warranted, even nephrotic syndrome to be efficacious than in pediatric
in patiens achieving remission, to recognize and treat adverse renal transplant recipients.
events and complications.
4. What statement on the therapeutic properties of
Mycophenolate Mofetil (MMF) is correct?

Multiple-choice questions (answers are a) MMF is not a therapeutic option in children with
provided following the reference list) steroid-sensitive nephrotic syndrome experiencing
frequent relapses or steroid dependency.
1. What statement on nephrotic syndrome in childhood is b) When using MMF as long-term immunosuppression,
correct? no further follow-up to detect potential side effects is
warranted.
a) Idiopathic nephrotic syndrome is by far the least fre- c) MMF is always beneficial in patients with steroid-
quent glomerular disease in childhood. resistant nephrotic syndrome.
b) About a half of patients with relapses have frequently d) The KDIGO expert working group has not recom-
relapsing nephrotic syndrome with one third to one mended MMF for patients with FRNS/SDNS
half of these being steroid dependent. experiencing steroid toxicity.
c) Azathioprine is the preferred steroid-sparing agent for e) MMF has immunosuppressive and antiproteinuric
treatment of nephrotic syndrome in childhood. effects.
d) Prolonged use of steroids for treatment of nephrotic 5. Critically reviewing published studies on Mycophenolate
syndrome in childhood is generally tolerated without Mofetil (MMF) for sustained remission in nephrotic syn-
any side effects. drome one has to consider that
e) There is broadly based consensus of steroid-sparing
treatment of nephrotic syndrome in childhood. a) there are important limitations to the interpretation and
2. Mycophenolic acid is comparability of published studies including study de-
sign, sample size, patient selection, clinical endpoints,
a) an ester prodrug of Mycophenolate Mofetil carry-over effects and duration of follow-up.
b) acts as an inhibitor of inosine monophosphate b) it is not possible to finally determine efficacy of MMF
dehydrogenase from the published uncontrolled studies,
c) counteracts the expansion of mesangial matrix c) it is currently unclear how long patients with fre-
d) has no systemic toxicity at all quently relapsing nephrotic syndrome, steroid-
e) seems to have an antiproteinuric effect dependent nephrotic syndrome or steroid-resistant ne-
phrotic syndrome should be treated with MMF.
Which statements are correct?
Which statements are correct?
a) Statements 1 and 2 are correct
b) All statements are correct a) All of the above statements are correct.
c) Statements 2, 3 and 5 are correct b) None of the above statements is correct.
d) Statements 2 to 5 are correct c) Only statement 1 is correct.
e) Statements 2 and 5 are correct d) Statements 2 and 3 are correct.
e) Statements 1 and 3 are correct.
3. What statement on pharmacokinetics of mycophenolic
acid is false? 6. What statement on principal limitations of clinical trials in
patients with relapsing nephrotic syndrome is correct?
a) There is a poor correlation between MMF dose and MPA
exposure. a) All steroid-sparing drugs used for treatment of the
b) The pharmacologically inactive 7-O-MPA glucuronide nephrotic syndrome have only weak immunosuppres-
(MPAG) is the major metabolite of MPA. sive properties.
c) MPA exhibits significant inter- and intra-patient variabil- b) The idiopathic nephrotic syndrome may follow its
ity in drug pharmacokinetics. natural course and may resolve with time. Treatment
d) Therapeutic drug monitoring of Mycophenolate Mofetil effects may thus be falsely attributed to a particular
plays no role in children with nephrotic syndrome. medication.
Pediatr Nephrol

c) Pharmacokinetics of mycophenolic acid do not show Mycophenolate mofetil reduces myofibroblast infiltration and col-
lagen III deposition in rat remnant kidney. Kidney Int 58:51–61
large inter- and intra-individual variability.
14. Cheng CC, Lee YF, Lan JL, Wu MJ, Hsieh TY, Lin NN, Wang JM,
d) The clinical response to steroids is always constant. Chiu YT (2013) Mycophenolate mofetil alleviates lupus nephritis
e) The disease affects mainly girls, and results of all through urokinase receptor signaling in a mice model. Lupus 22:
studies may be biased by a preponderance of female 554–561
15. Hackl A, Ehren R, Weber LT (2017) Effect of mycophenolic
participants.
acid in experimental, nontransplant glomerular diseases: new
mechanisms beyond immune cells. Pediatr Nephrol 32:1315–
Compliance with ethical standards 1322
16. Weber SC, Uhlenberg B, Raile K, Querfeld U, Muller D (2011)
Conflict of interest Uwe Querfeld declares that he has no conflict of Polyoma virus-associated progressive multifocal leukoencephalopathy
interest. Lutz T. Weber has received financial support (travel grants) from after renal transplantation: regression following withdrawal of myco-
Astellas. phenolate mofetil. Pediatr Transplant 15:E19–E24
17. Neff RT, Hurst FP, Falta EM, Bohen EM, Lentine KL,
Dharnidharka VR, Agodoa LY, Jindal RM, Yuan CM, Abbott KC
(2008) Progressive multifocal leukoencephalopathy and use of my-
References cophenolate mofetil after kidney transplantation. Transplantation
86:1474–1478
18. Perez-Aytes A, Marin-Reina P, Boso V, Ledo A, Carey JC, Vento M
1. Hodson EM, Willis NS, Craig JC (2008) Non-corticosteroid treat- (2017) Mycophenolate mofetil embryopathy: a newly recognized
ment for nephrotic syndrome in children. Cochrane Database Syst teratogenic syndrome. Eur J Med Genet 60:16–21
Rev:CD002290 19. Kemper MJ, Valentin L, van Husen M (2017) Difficult-to-treat
2. Pravitsitthikul N, Willis NS, Hodson EM, Craig JC (2013) Non- idiopathic nephrotic syndrome: established drugs, open questions
corticosteroid immunosuppressive medications for steroid-sensitive and future options. Pediatr Nephrol. https://doi.org/10.1007/
nephrotic syndrome in children. Cochrane Database Syst Rev: s00467-017-3780-7
CD002290 20. Colucci M, Carsetti R, Cascioli S, Casiraghi F, Perna A, Rava L,
3. Group KDIGOKGW (2012) KDIGO clinical practice guideline for Ruggiero B, Emma F, Vivarelli M (2016) B cell reconstitution after
glomerulonephritis. Kidney Int Suppl 2:139–274 rituximab treatment in idiopathic nephrotic syndrome. J Am Soc
4. Querfeld U, Dötsch J, Gellermann J, Hoyer P, Kemper M, Nephrol 27:1811–1822
Latta K, Tönshoff B, Weber LT, Rascher W (2017) 21. Dorresteijn EM, Kist-van Holthe JE, Levtchenko EN, Nauta J, Hop
Diagnosis and therapy of idiopathic nephrotic syndrome in WC, van der Heijden AJ (2008) Mycophenolate mofetil versus
childhood. Summary of the S2e guideline AWMF registry cyclosporine for remission maintenance in nephrotic syndrome.
number 166-001, headed by the German Society for Pediatr Nephrol 23:2013–2020
Pediatric Nephrology. Monatsschr Kinderheilkd 165:997– 22. Gellermann J, Weber L, Pape L, Tonshoff B, Hoyer P, Querfeld U
1004 (2013) Mycophenolate mofetil versus cyclosporin A in children
5. Lombel RM, Gipson DS, Hodson EM, Kidney Disease: Improving with frequently relapsing nephrotic syndrome. J Am Soc Nephrol
Global O (2013) Treatment of steroid-sensitive nephrotic syn- 24:1689–1697
drome: new guidelines from KDIGO. Pediatr Nephrol 28:415–426 23. Sinha A, Gupta A, Kalaivani M, Hari P, Dinda AK, Bagga A (2017)
6. Allison AC, Eugui EM (2005) Mechanisms of action of mycophe- Mycophenolate mofetil is inferior to tacrolimus in sustaining remis-
nolate mofetil in preventing acute and chronic allograft rejection. sion in children with idiopathic steroid-resistant nephrotic syn-
Transplantation 80:S181–S190 drome. Kidney Int 92:248–257
7. Camici M, Micheli V, Ipata PL, Tozzi MG (2010) Pediatric neuro- 24. B a g g a A , H a r i P, M o u d g i l A , J o r d a n S C ( 2 0 0 3 )
logical syndromes and inborn errors of purine metabolism. Mycophenolate mofetil and prednisolone therapy in children
with steroid-dependent nephrotic syndrome. Am J Kidney
Neurochem Int 56:367–378
Dis 42:1114–1120
8. Rovin BH (2012) Lupus nephritis: guidelines for lupus nephritis–
25. Novak I, Frank R, Vento S, Vergara M, Gauthier B, Trachtman H
more recommendations than data? Nat Rev Nephrol 8:620–621
(2005) Efficacy of mycophenolate mofetil in pediatric patients with
9. Conti F, Ceccarelli F, Perricone C, Massaro L, Cipriano E, Pacucci steroid-dependent nephrotic syndrome. Pediatr Nephrol 20:1265–1268
VA, Truglia S, Miranda F, Morello F, Alessandri C, Spinelli FR, 26. Hogg RJ, Fitzgibbons L, Bruick J, Bunke M, Ault B, Baqi N,
Valesini G (2014) Mycophenolate mofetil in systemic lupus erythe- Trachtman H, Swinford R (2006) Mycophenolate mofetil in chil-
matosus: results from a retrospective study in a large monocentric dren with frequently relapsing nephrotic syndrome: a report from
cohort and review of the literature. Immunol Res 60:270–276 the Southwest Pediatric Nephrology Study Group. Clin J Am Soc
10. Ziswiler R, Steinmann-Niggli K, Kappeler A, Daniel C, Marti HP Nephrol 1:1173–1178
(1998) Mycophenolic acid: a new approach to the therapy of exper- 27. Okada M, Sugimoto K, Yagi K, Yanagida H, Tabata N, Takemura T
imental mesangial proliferative glomerulonephritis. J Am Soc (2007) Mycophenolate mofetil therapy for children with intractable
Nephrol 9:2055–2066 nephrotic syndrome. Pediatr Int 49:933–937
11. Hauser IA, Renders L, Radeke HH, Sterzel RB, Goppelt-Struebe M 28. Fujinaga S, Ohtomo Y, Umino D, Takemoto M, Shimizu T,
(1999) Mycophenolate mofetil inhibits rat and human mesangial Yamashiro Y, Kaneko K (2007) A prospective study on the use of
cell proliferation by guanosine depletion. Nephrol Dial Transplant mycophenolate mofetil in children with cyclosporine-dependent
14:58–63 nephrotic syndrome. Pediatr Nephrol 22:71–76
12. Dubus I, Vendrely B, Christophe I, Labouyrie JP, Delmas Y, Bonnet 29. Afzal K, Bagga A, Menon S, Hari P, Jordan SC (2007) Treatment
J, Combe C (2002) Mycophenolic acid antagonizes the activation with mycophenolate mofetil and prednisolone for steroid-
of cultured human mesangial cells. Kidney Int 62:857–867 dependent nephrotic syndrome. Pediatr Nephrol 22:2059–2065
13. Badid C, Vincent M, McGregor B, Melin M, Hadj-Aissa A, 30. Fujinaga S, Ohtomo Y, Hirano D, Nishizaki N, Someya T, Ohtsuka
Veysseyre C, Hartmann DJ, Desmouliere A, Laville M (2000) Y, Kaneko K, Shimizu T (2009) Mycophenolate mofetil therapy for
Pediatr Nephrol

childhood-onset steroid dependent nephrotic syndrome after long- steroid-dependent nephrotic syndrome. Pediatr Nephrol 26:
term cyclosporine: extended experience in a single center. Clin 1823–1828
Nephrol 72:268–273 49. Gellermann J, Ehrich JH, Querfeld U (2012) Sequential mainte-
31. Baudouin V, Alberti C, Lapeyraque AL, Bensman A, Andre JL, nance therapy with cyclosporin A and mycophenolate mofetil for
Broux F, Cailliez M, Decramer S, Niaudet P, Deschenes G, Jacqz- sustained remission of childhood steroid-resistant nephrotic syn-
Aigrain E, Loirat C (2012) Mycophenolate mofetil for steroid- drome. Nephrol Dial Transplant 27:1970–1978
dependent nephrotic syndrome: a phase II Bayesian trial. Pediatr 50. Nikibakhsh AA, Mahmoodzadeh H, Karamyyar M, Hejazi S,
Nephrol 27:389–396 Noroozi M, Macooie AA (2011) Treatment of steroid and
32. Banerjee S, Pahari A, Sengupta J, Patnaik SK (2013) Outcome of cyclosporine-resistant idiopathic nephrotic syndrome in children.
severe steroid-dependent nephrotic syndrome treated with myco- Int J Nephrol 2011:930965
phenolate mofetil. Pediatr Nephrol 28:93–97 51. Hibino S, Uemura O, Nagai T, Yamakawa S, Iwata N, Ito H,
33. Wang J, Mao J, Chen J, Fu H, Shen H, Zhu X, Liu A, Shu Nakano M, Tanaka K (2015) Three year outcome of childhood
Q, Du L (2016) Evaluation of mycophenolate mofetil or idiopathic nephrotic syndrome under a unified immunosuppressive
tacrolimus in children with steroid sensitive but frequently protocol. Pediatr Int 57:85–91
relapsing or steroid-dependent nephrotic syndrome. 52. Wu B, Mao J, Shen H, Fu H, Wang J, Liu A, Gu W, Shu
Nephrology (Carlton) 21:21–27 Q, Du L (2015) Triple immunosuppressive therapy in
34. Jellouli M, Fitouhi S, Abidi K, Hammi Y, Naija O, Zarrouk C, steroid-resistant nephrotic syndrome children with tacrolimus
Gargah T (2016) Mycophenolate mofetil in treatment of childhood resistance or tacrolimus sensitivity but frequently relapsing.
steroid-dependent nephrotic syndrome. Tunis Med 94:221–225 Nephrology (Carlton) 20:18–24
35. Basu B, Babu BG, Mahapatra TK (2017) Long-term efficacy and 53. Kim J, Patnaik N, Chorny N, Frank R, Infante L, Sethna C
safety of common steroid-sparing agents in idiopathic nephrotic (2014) Second-line immunosuppressive treatment of child-
children. Clin Exp Nephrol 21:143–151 hood nephrotic syndrome: a single-center experience.
36. Barletta GM, Smoyer WE, Bunchman TE, Flynn JT, Nephron Extra 4:8–17
Kershaw DB (2003) Use of mycophenolate mofetil in 54. Hocker B, Weber LT, Bunchman T, Rashford M, Tonshoff B,
steroid-dependent and -resistant nephrotic syndrome. Tricontinental MMFSSG (2005) Mycophenolate mofetil suspen-
Pediatr Nephrol 18:833–837 sion in pediatric renal transplantation: three-year data from the
37. Montane B, Abitbol C, Chandar J, Strauss J, Zilleruelo G (2003) tricontinental trial. Pediatr Transplant 9:504–511
Novel therapy of focal glomerulosclerosis with mycophenolate and
55. Weber LT (2015) Therapeutic drug monitoring in pediatric renal
angiotensin blockade. Pediatr Nephrol 18:772–777
transplantation. Pediatr Nephrol 30:253–265
38. Gellermann J, Querfeld U (2004) Frequently relapsing nephrotic
56. Filler G, Alvarez-Elias AC, McIntyre C, Medeiros M (2017) The
syndrome: treatment with mycophenolate mofetil. Pediatr Nephrol
compelling case for therapeutic drug monitoring of mycophenolate
19:101–104
mofetil therapy. Pediatr Nephrol 32:21–29
39. Ulinski T, Dubourg L, Said MH, Parchoux B, Ranchin B, Cochat P
(2005) Switch from cyclosporine A to mycophenolate mofetil in 57. Barraclough KA, Lee KJ, Staatz CE (2010) Pharmacogenetic influ-
nephrotic children. Pediatr Nephrol 20:482–485 ences on mycophenolate therapy. Pharmacogenomics 11:369–390
40. Mendizabal S, Zamora I, Berbel O, Sanahuja MJ, Fuentes J, Simon 58. Kuypers DR, Le Meur Y, Cantarovich M, Tredger MJ, Tett
J (2005) Mycophenolate mofetil in steroid/cyclosporine-dependent/ SE, Cattaneo D, Tonshoff B, Holt DW, Chapman J, Gelder
resistant nephrotic syndrome. Pediatr Nephrol 20:914–919 T, Transplantation Society Consensus Group on TDMoMPA
41. de Mello VR, Rodrigues MT, Mastrocinque TH, Martins SP, de (2010) Consensus report on therapeutic drug monitoring of
Andrade OV, Guidoni EB, Scheffer DK, Martini Filho D, mycophenolic acid in solid organ transplantation. Clin J Am
Toporovski J, Benini V (2010) Mycophenolate mofetil in children Soc Nephrol 5:341–358
with steroid/cyclophosphamide-resistant nephrotic syndrome. 59. Sobiak J, Resztak M, Ostalska-Nowicka D, Zachwieja J,
Pediatr Nephrol 25:453–460 Gasiorowska K, Piechanowska W, Chrzanowska M (2015)
42. Li Z, Duan C, He J, Wu T, Xun M, Zhang Y, Yin Y (2010) Monitoring of mycophenolate mofetil metabolites in children
Mycophenolate mofetil therapy for children with steroid-resistant with nephrotic syndrome and the proposed novel target
nephrotic syndrome. Pediatr Nephrol 25:883–888 values of pharmacokinetic parameters. Eur J Pharm Sci 77:
43. Hassan AV, Sinha MD, Waller S (2013) A single-centre retrospective 189–196
study of the safety and efficacy of mycophenolate mofetil in children 60. Tong K, Mao J, Fu H, Shen H, Liu A, Shu Q, Du L (2016) The
and adolescents with nephrotic syndrome. Clin Kidney J 6:384–389 value of monitoring the serum concentration of mycophenolate mo-
44. Al-Akash S, Al-Makdama A (2005) Mycophenolate mofetil in chil- fetil in children with steroid-dependent/frequent relapsing nephrotic
dren with steroid-dependent and/or frequently relapsing nephrotic syndrome. Nephron 132:327–334
syndrome. Ann Saudi Med 25:380–384 61. Hackl A, Cseprekal O, Gessner M, Liebau MC, Habbig S, Ehren R,
45. Gargah TT, Lakhoua MR (2011) Mycophenolate mofetil in treat- Muller C, Taylan C, Dotsch J, Weber LT (2016) Mycophenolate
ment of childhood steroid-resistant nephrotic syndrome. J Nephrol mofetil therapy in children with idiopathic nephrotic syndrome:
24:203–207 does therapeutic drug monitoring make a difference? Ther Drug
46. Nickavar A, Safarzadeh AE, Sotoudeh K, Otukesh H, Monit 38:274–279
Hooman N (2012) Mycophenolate mofetil for treatment of 62. Weber LT, Shipkova M, Armstrong VW, Wagner N, Schutz E,
idiopathic nephrotic syndrome in children. Iran J Kidney Dis Mehls O, Zimmerhackl LB, Oellerich M, Tonshoff B (2002) The
6:346–349 pharmacokinetic-pharmacodynamic relationship for total and free
47. Fujinaga S, Someya T, Watanabe T, Ito A, Ohtomo Y, Shimizu T, mycophenolic acid in pediatric renal transplant recipients: a report
Kaneko K (2013) Cyclosporine versus mycophenolate mofetil for of the german study group on mycophenolate mofetil therapy. J Am
maintenance of remission of steroid-dependent nephrotic syndrome Soc Nephrol 13:759–768
after a single infusion of rituximab. Eur J Pediatr 172:513–518 63. Filler G, Lepage N, Delisle B, Mai I (2001) Effect of cyclosporine
48. Ito S, Kamei K, Ogura M, Sato M, Fujimaru T, Ishikawa T, on mycophenolic acid area under the concentration-time curve in
Udagawa T, Iijima K (2011) Maintenance therapy with my- pediatric kidney transplant recipients. Ther Drug Monit 23:514–
cophenolate mofetil after rituximab in pediatric patients with 519
Pediatr Nephrol

64. Saint-Marcoux F, Guigonis V, Decramer S, Gandia P, Ranchin B, Bensman A, Azougagh S, Jacqz-Aigrain E (2010) Population phar-
Parant F, Bessenay L, Libert F, Harambat J, Bouchet S, Broux F, macokinetics and pharmacogenetics of mycophenolic acid follow-
Compagnon P, Marquet P (2011) Development of a Bayesian estimator ing administration of mycophenolate mofetil in de novo pediatric
for the therapeutic drug monitoring of mycophenolate mofetil in chil- renal-transplant patients. J Clin Pharmacol 50:1280–1291
dren with idiopathic nephrotic syndrome. Pharmacol Res 63:423–431
65. Zhao W, Fakhoury M, Deschenes G, Roussey G, Brochard K, Answers
Niaudet P, Tsimaratos M, Andre JL, Cloarec S, Cochat P, 1. b; 2. c; 3. d; 4. e; 5. a; 6. b

Vous aimerez peut-être aussi