ArticlePDF Available

Treatment of Resistant Glomerular Diseases with Adrenocorticotropic Hormone Gel: A Prospective Trial

Authors:

Abstract and Figures

Adrenocorticotropic hormone (ACTH) has shown promising results in glomerular diseases resistant to conventional therapies, but the reported data have solely been from retrospective, observational studies. In this prospective, open-label study (NCT01129284), 15 subjects with resistant glomerular diseases were treated with ACTH gel (80 units subcutaneously twice weekly) for 6 months. Resistant membranous nephropathy (MN), minimal change disease (MCD), and focal segmental glomerulosclerosis (FSGS) were defined as failure to achieve sustained remission of proteinuria off immunosuppressive therapy with at least 2 treatment regimens; resistant IgA nephropathy was defined as >1 g/g urine protein:creatinine ratio despite maximally tolerated RAAS blockade. Remission was defined as stable or improved renal function with ≥50% reduction in proteinuria to <0.5 g/g (complete remission) or 0.5-3.5 g/g (partial remission). The study included 5 subjects with resistant idiopathic MN, 5 subjects with resistant MCD (n = 2)/FSGS (n = 3), and 5 subjects with resistant IgA nephropathy. Two resistant MN subjects achieved partial remission on ACTH therapy, although 3 achieved immunologic remission of disease (PLA(2)R antibody disappeared by 4 months of therapy). One subject with resistant FSGS achieved complete remission on ACTH; one subject with resistant MCD achieved partial remission but relapsed within 4 weeks of stopping ACTH. Two subjects with resistant IgA nephropathy demonstrated >50% reductions in proteinuria while on ACTH, with proteinuria consistently <1 g/g by 6 months. Three of 15 subjects reported significant steroid-like adverse effects with ACTH, including weight gain and hyperglycemia, prompting early termination of therapy without any clinical response. ACTH gel is a promising treatment for resistant glomerular diseases and should be studied further in controlled trials against currently available therapies for resistant disease.
Content may be subject to copyright.
Fax +41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Original Report: Patient-Oriented, Translational Research
Am J Nephrol 2012;36:58–67
DOI: 10.1159/000339287
Treatment of Resistant Glomerular
Diseases with Adrenocorticotropic
Hormone Gel: A Prospective Trial
Andrew S. Bomback
a
Pietro A. Canetta
a
Laurence H. Beck Jr.
b
Rivka Ayalon
b
Jai Radhakrishnan
a
Gerald B. Appel
a
a Division of Nephrology, Department of Medicine, Columbia University Medical Center, New York, N.Y. , and
b Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston, Mass. , USA
phropathy. Two resistant MN subjects achieved partial re-
mission on ACTH therapy, although 3 achieved immunolog-
ic remission of disease (PLA
2
R antibody disappeared by 4
months of therapy). One subject with resistant FSGS achieved
complete remission on ACTH; one subject with resistant
MCD achieved partial remission but relapsed within 4 weeks
of stopping ACTH. Two subjects with resistant IgA nephrop-
athy demonstrated 1 50% reductions in proteinuria while on
ACTH, with proteinuria consistently ! 1 g/g by 6 months.
Three of 15 subjects reported significant steroid-like adverse
effects with ACTH, including weight gain and hyperglyce-
mia, prompting early termination of therapy without any
clinical response. Conclusions: ACTH gel is a promising
treatment for resistant glomerular diseases and should be
studied further in controlled trials against currently available
therapies for resistant disease.
Copyright © 2012 S. Karger AG, Bas el
Introduction
Patients with idiopathic glomerular diseases often re-
quire immunosuppression to achieve remissions of dis-
ease, defined by resolution of proteinuria with stable or
improving renal function. Unfortunately, many patients
Key Words
Membranous nephropathy Minimal change disease
Focal segmental glomerulosclerosis IgA nephropathy
A b s t r a c t
B a c k g r o u n d : Adrenocorticotropic hormone (ACTH) has
shown promising results in glomerular diseases resistant to
conventional therapies, but the reported data have solely
been from retrospective, observational studies. Methods: In
this prospective, open-label study (NCT01129284), 15 sub-
jects with resistant glomerular diseases were treated with
ACTH gel (80 units subcutaneously twice weekly) for 6
months. Resistant membranous nephropathy (MN), minimal
change disease (MCD), and focal segmental glomeruloscle-
rosis (FSGS) were defined as failure to achieve sustained re-
mission of proteinuria off immunosuppressive therapy with
at least 2 treatment regimens; resistant IgA nephropathy was
defined as 1 1 g/g urine protein:creatinine ratio despite max-
imally tolerated RAAS blockade. Remission was defined as
stable or improved renal function with 6 50% reduction in
proteinuria to ! 0.5 g/g (complete remission) or 0.5–3.5 g/g
(partial remission). Results: The study included 5 subjects
with resistant idiopathic MN, 5 subjects with resistant MCD
(n = 2)/FSGS (n = 3), and 5 subjects with resistant IgA ne-
Recei ved: February 28, 2012
Accepted: May 2, 2012
Published onlin e: June 19, 2012
Nephrolo
gy
American Journal of
Andrew S. Bomback, MD, MPH
Department of Med icine, Division of Nephrology
Columbia University Medical Center
622 West 168t h Street, PH 4-124, New York, NY 10032 (USA)
Tel. +1 212 305 0320, E-Ma il asb68
@ columbia.edu
© 2012 S. Ka rger AG, Basel
0250–8095/12/0361–0058$38.00/0
Accessible online at:
www.karger.com/ajn
ACTH for Resistant Glomerular Diseases Am J Nephrol 2012;36:58–67
59
do not respond to first- or second-line treatment choices
or relapse once immunosuppression has been reduced or
discontinued. For example, while up to 90% of adults
with minimal change disease (MCD) will respond to ini-
tial therapy with prednisone, approximately 40% of these
same patients will relapse within 6 months and require
further immunosuppression
[1] . First-line therapies for
diseases such as idiopathic membranous nephropathy
(MN) and focal segmental glomerulosclerosis (FSGS)
produce substantially lower initial response rates and are
marked by similarly high relapse rates
[2–5] . Thus, physi-
cians who treat these patients commonly employ second-,
third- and even fourth-line therapies in attempting to
achieve a sustained remission. New treatment options are
always in demand.
In the last two decades, adrenocorticotropic hormone
(ACTH) has re-emerged as a potential treatment option
for patients with idiopathic nephrotic syndrome, with
benefit shown in a variety of disease states ranging from
idiopathic MN and FSGS to diabetic nephropathy and
hereditary nephritis. The mechanism of action by which
ACTH ameliorates proteinuria remains unclear. Pro-
posed mechanisms include stimulation of endogenous
steroid production, activation of melanocortin receptors
on inflammatory cells (including B cells, T cells, and an-
tigen-presenting cells), and direct binding to melanocor-
tin receptors on the podocyte.
Virtually all of the experience using ACTH for ne-
phrotic syndrome comes from retrospective, observa-
tional case series of heterogeneous patient populations,
including patients who received ACTH as first-line ther-
apy and patients who were treated after failing 2, 3, or 4
prior immunomodulatory regimens
[6–10] . To date, only
one prospective study of ACTH has been reported, a ran-
domized trial comparing ACTH and cyclophosphamide
as initial therapy in 32 patients with idiopathic MN
[11] .
Given this lack of prospective data, particularly among
patients with resistant glomerular diseases, the popula-
tion in which ACTH is almost exclusively used in the
United States
[9] , we performed a prospective, open-la -
bel study of ACTH in patients with resistant forms of
the most common idiopathic glomerular diseases: MN,
FSGS, MCD and IgA nephropathy.
M e t h o d s
S u b j e c t s
This open-label, nonblinded, efficacy and safety study of
ACTH gel (H.P. Acthar Gel, repository corticotropin injection,
Questcor Pharmaceuticals, Inc., Hayward, Calif., USA), a long-
acting formulation of full-sequence ACTH(1–39) that includes
other pro-opiomelanocortin peptides, consisted of 15 subjects
with resistant glomerular diseases: 5 subjects with resistant MN,
5 subjects with MCD or FSGS, and 5 subjects with resistant IgA
nephropathy. Resistant MN was defined as failure to achieve sus-
tained remission with at least 2 prior immunosuppressive regi-
mens, resistant MCD/FSGS was defined as failure to achieve sus-
tained remission with corticosteroids and at least 1 other immu-
nosuppressive regimen, and resistant IgA nephropathy was
defined as 1 g/g urine protein:creatinine ratio despite stable and
maximally tolerated doses of renin-angiotensin-aldosterone sys-
tem (RAAS) blockade. Exclusion criteria were age ^ 18 years, e s-
timated GFR ! 30 ml/min/1.73 m
2
, use of rituximab or other
monoclonal antibody within 6 months of the trial, use of cyclo-
phosphamide within 3 months of the trial, ongoing pregnancy or
breastfeed ing, refusa l to use birth control if ch ildbearing age, ot h-
er renal diseases (e.g. diabetes, renal vascular disease) that would
interfere with interpretation of study results, and known contra-
indications to the use of ACTH gel (scleroderma, osteoporosis,
system ic fu ngal infect ions, ocu lar herp es simplex, recent surgery,
history of or the presence of a peptic ulcer, congestive heart fail-
ure, uncontrolled hypertension, and allergies to pork or pork
products). All subjects signed an informed consent prior to en-
rollment. The study was approved by the Institutional Review
Board of Columbia University Medical Center and registered on
clinicaltrials.gov (NCT01129284).
Treatment Regimen and Evaluations
Subjects were treated with ACTH gel for 24 weeks, dosed at 40
units twice weekly subcutaneously for 2 weeks, then 80 units
twice weekly subcutaneously afterwards. Twice weekly treat-
ments were given at the study site for weeks 1 through 3, during
which time subjects received instructions for home administra-
tion. Medication could be injected in the arm, abdomen, or thigh.
Starting at week 4, subjects were given one treatment at the study
site and administered the other treatment at home, maintaining
the same t wice weekly dosing schedule. Subjects who did not wish
to self-administer the treatment were given the option to receive
all treatments at the study center. The twice weekly injections
were spaced out 72–96 h apart. No changes were made to subjects’
RAAS-blocking medications or cholesterol-lowering medica-
tions during the treatment period unless clinically indicated (e.g.
hyperkalemia). Subjects who were on calcineurin inhibitors were
weaned off this therapy during the first 12 weeks of ACTH ther-
apy.
Laboratory measurements were performed week ly for the first
4 weeks of the study period, and then every 4 weeks thereafter
until the conclusion of the study. These measurements included
basic metabolic panel, complete blood count, hepatic function
panel, lipid prof ile, spot uri ne protein:creatini ne ratio, serum cor-
tisol (reference range 5.025.0 g/dl), and serum ACTH (refer-
ence range 7–69 pg/ml). Blood and urine samples were collected
immediately prior to ACTH injections. Baseline proteinuria was
calculated by the average of the first 3 weeks’ urine protein:creati-
nine ratio (U PCR) values. Stored serum s amples from the subjects
with MN were sent, after completion of the trial, to Boston Uni-
versity for antibody testing against the M-type phospholipase A
2
receptor (PLA
2
R) to serve as a serologic marker of disease activ-
ity. The details of this immunoassay have been described else-
where
[12 ] .
Bomback /Canetta /BeckJr. /Aya lon /
Radhakrishnan
/Ap pe l
Am J Nephrol 2012;36:58–67
60
O u t c o m e s
The primary outcome for subjects with nephrotic syndrome
was remission st atus at the completion of ACTH therapy. Complete
remission was def ined as stable or improved renal function (creati-
nine ^ 125% of baseline) with final UPCR ! 0.500 g/g. Partial re-
mission was defined as stable or improved renal function with
6 50% reduction in UPCR to 0.500–3.500 g/g. All subjects failing
to meet these criteria were deemed nonresponders. The primary
outcome for subjects with IgA nephropathy was percent reduction
in proteinuria from baseline at the completion of ACTH therapy.
Se con dar y outc omes in cluded cha nge in s erum a lbu mi n, c han ge i n
total cholesterol a nd LDL cholesterol, and adverse effect s of therapy.
Statistical Analysis
Given the small number of subjects and the pilot design of this
study, no formal statistical analysis was performed. Instead, the
data are presented descriptively.
R e s u l t s
Fifteen subjects were included in this open-label pilot
study of ACTH gel for resistant glomerular diseases ( ta-
ble1 ). The five subjects with MN all had impaired renal
function at baseline (serum creatinine range 1.6–2.9 mg/
dl). The most commonly failed prior therapies in this
group were mycophenolate mofetil (MMF, n = 4) and cal-
cineurin inhibitors (n = 3). Two subjects had previously
been treated with cyclophosphamide. The 5 subjects with
resistant FSGS or MCD had preserved renal function at
baseline (serum creatinine range 0.6–1.2 mg/dl) and, per
protocol, all had failed a traditional course of oral corti-
costeroids. All of these subjects failed at least one course
of calcineurin inhibitor therapy, and 3 had previously
been on MMF without sustained remission. No subject in
this group had previously been treated with cyclophos-
phamide. The five subjects with IgA nephropathy includ-
ed 2 subjects with preserved kidney function (eGFR 1 60
ml/min/1.73 m
2
) and 3 subjects with impaired kidney
function; all were on stable doses of RAAS blockade. The
3 subjects with IgA nephropathy and reduced eGFR had
previously been treated with a 6-month course of corti-
costeroids without response.
Two of the 5 subjects with MN achieved partial remis-
sion over the 6-month course of ACTH therapy ( fig. 1 ;
Table 1. Baseline characteristics of study participants
Age Race/
ethnicity
Sex Months
from
Previous immunosuppressive therapy Pre-ACTH
Screat
Pre-ACTH eGFR
ml/min/1.73 m2
Pre-ACTH
UPCR
diagnosis mg/dl g/g
MN
Subject 1 68 white male 78 steroids, cyclosporine, tacrolimus 1.6 44 6.70
Subject 2 50 Af-Am female 108 MMF, tacrolimus, cyclosporine,
rituximab
2.0 33 3.05
Subject 3 68 Asian male 49 steroids, MMF, cyclophosphamide 2.2 34 3.80
Subject 4 58 white male 58 cyclosporine, MMF 1.7 43 2.23
Subject 5 71 white male 12 MMF, cyclophosphamide 2.9a 21
a 12.46
FSGS/MCD
Subject 6 29 Hispanic female 120 steroids, MMF, rituximab, tacrolimus 0.7 117 4.76
Subject 7 30 white female 57 steroids, tacrolimus 0.6 123 3.16
Subject 8 41 white male 87 steroids, tacrolimus, cyclosporine,
MMF
1.2 75 1.94
Subject 9 35 white male 99 steroids, cyclosporine 0.8 116 1.96
Subject 10 66 white male 128 steroids, cyclosporine, MMF 1.0 78 1.65
IgA nephropathy
Subject 11 37 Hispanic female 10 steroids 1.7 38 1.95
Subject 12 46 white male 31 steroids, MMF 2.4 31 1.59
Subject 13 55 Asian female 1 none 0.8 83 0.61b
Subject 14 36 Asian male 84 steroids 2.7 29 4.12
Subject 15 35 Asian male 7 none 1.0 97 1.51
Af-Am = African-American; eGFR = estimated glomerular filtration rate (CKD EPI formula); S
creat
= serum creatinine.
a At screening visit, subject 5’s serum creatinine was 2.1 and eGFR was 31 ml/min/1.73 m2.
b At screening visit, subject 13’s UPCR was 1.543 mg/g.
ACTH for Resistant Glomerular Diseases Am J Nephrol 2012;36:58–67
61
table2 ). Subject 4, whose creatinine levels fell from 1.7 to
1.2 mg/dl, albumin levels rose from 2.9 to 4.0 g/dl and
UPCR fell from 2.23 to 0.84 g/g, proceeded to have a sig-
nificant decline in proteinuria after ACTH was with-
drawn, achieving complete remission of proteinuria
within 3 months of stopping ACTH. He continues to
demonstrate sustained complete remission 12 months af-
ter his last dose of ACTH, with creatinine stable at 1.3 mg/
d l a nd UP CR 0.07 g/g . A nt ib od ie s t o t he PL A
2
R were con-
sistently negative for this subject. Subject 5 experienced
both clinical and immunologic remission of disease while
on ACTH: creatinine fell from 2.9 to 1.4 mg/dl, albumin
rose from 2.1 to 3.4 g/dl, and UPCR fell from 12.46 to 3.14
g/g, while antibodies to the PLA
2
R went from strongly
positive to negative. Surveillance laboratories done 10
months after stopping ACTH showed that, clinically, this
remission was maintained (UPCR was 0.87 g/g) despite
reappearance of anti-PLA
2
R antibodies. Testing for anti-
bodies to the PLA
2
R suggested that subjects 2 and 3
achieved an immunologic remission of disease (antibod-
ies went from strongly positive to negative) during treat-
ment with ACTH despite no apparent improvement in
clinical parameters. Subject 2 remains PLA
2
R antibody
negative more than one year from her last dose of ACTH
with proteinuria ranging from 3.0 to 4.5 g/day; subject 3
was lost to long-term follow-up after the study. As a
group, the 5 subjects with MN went from a pre-ACTH
median (range) proteinuria of 3.80 mg/g (2.2312.46) to
a post-ACTH median (range) proteinuria of 3.79 mg/g
(1.37–10. 50).
One subject with MCD (subject 7) and 1 subject with
FSGS (subject 8) achieved partia l remission of proteinuria
during ACTH therapy ( fig.2 ; table3 ). Subject 7 demon-
strated a drop in UPCR from peak 3.16 to 0.78 g/g with
albumin rising from nadir 3.3 to 3.7 g/dl during treat-
ment; however, UPCR rose to 1 3.50 g/g 4 weeks after
stopping ACTH. Subject 8 demonstrated a drop in UPCR
f rom pe ak 3. 27 to 0.43 g/g wi th album in ri si ng fr om na di r
3.2 to 3.9 g/dl during treatment. At 8 months since dis-
continuation of ACTH, his UPCR was 0.68 g/g. Subjects
6 (MCD) and 9 (FSGS), per protocol, were weaned off
calcineurin inhibitor therapy during the first 3 months
of their ACTH treatment regimens; both subjects saw
marked rises in proteinuria with declining serum albu-
min level, and both subjects remain calcineurin inhibi-
0
2
4
6
8
10
12 UPCR pre-ACTH (g/g)
14
Subject 1 Subject 2 Subject 3 Subject 4 Subject 5
UPCR post-ACTH (g/g)
0
2
4
6
8
10
UPCR pre-ACTH (g/g)
12
Subject 6 Subject 7 Subject 8 Subject 9 Subject 10
UPCR post-ACTH (g/g)
0
1
2
3
4
UPCR pre-ACTH (g/g)
5
Subject 11 Subject 12 Subject 13 Subject 14 Subject 15
UPCR post-ACTH (g/g)
F i g . 1 . Changes in UPCR over a 6-month ACTH treatment period
in subjects with MN. Detailed laboratory values over this time
period are presented in table2. Subjects 1 and 2 withdrew early
from the study.
F i g . 2 . Changes in UPCR over a 6-month ACTH treatment period
in subjects with MCD or FSGS. Detailed laboratory values over
this time period are presented in table3.
F i g . 3 . Changes in UPCR over a 6-month ACTH treat ment period
in subje cts with Ig A ne phr opa thy. Det ai led la bor ato ry value s ov er
this time period are presented in table4. Subject 14 withdrew ear-
ly from the study.
Bomback /Canetta /BeckJr. /Aya lon /
Radhakrishnan
/Ap pe l
Am J Nephrol 2012;36:58–67
62
tor-dependent approximately 1 year after therapy. As a
group, the 5 subjects with MCD or FSGS went from a pre-
ACTH median (range) proteinuria of 1.96 mg/g (1.65
4.76) to a post-ACTH median (range) proteinuria of 1.93
mg/g (0.43–11.35).
All but 1 subject with IgA nephropathy showed reduc-
tions in proteinuria during ACTH therapy ( fig.3 ; table4 ).
All subjects were on stable RAAS-blocking medications
for at least 6 months prior to enrollment, at highest doses
tolerated by blood pressure and/or serum potassium
(losartan 25 mg daily for subject 11, losartan 100 mg dai-
ly for subject 12, ramipril 10 mg daily + spironolactone 25
mg daily for subject 13, irbesartan 75 mg daily for subject
14, and enalapril 5 mg daily for subject 15). Subjects 12
and 13 demonstrated the largest reductions in UPCR
from 1.59 to 0.57 g/g (64%) and 0.61 to 0.21 g/g (66%), re-
spectively. These reductions have been sustained at 11
and 9 months, respectively, from last ACTH dose. Subject
11, whose UPCR fell from 1.95 to 1.22 g/g (38%) during
ACTH therapy, was unable to sustain this reduction in
proteinuria in the setting of discontinuing her RAAS-
blocking regimen to plan a pregnancy. Subject 15, whose
UPCR fell from 1.51 to 0.85 g/g during treatment, has
shown further declines in proteinuria (last UPCR 0.28
Table 2. Detailed results from 5 subjects with resistant MN
Baseline Week 4 Week 8 Week 12 Week 16 Week 20 Week 24
Subject 1
Scr, mg/dl 1.6 1.5 1.5 1.4
Salb, g/dl 3.2 3.0 2.5 2.6
UPCR, g/g 6.70 9.11 8.71 10.50
Cholesterol, mg/dl 148 171 177 151
Cortisol, g/dl 11.7 14.1 8.3 9.7
ACTH, pg/ml 11 13 15 8
Subject 2
Scr, mg/dl 2 1.8 1.9 1.7 1.8
Salb, g/dl 4.1 3.6 3.3 3.4 3.7
UPCR, g/g 3.05 2.88 3.58 3.79 3.75
Cholesterol, mg/dl 196 166 169 196
Cortisol, g/dl 13.4 10.1 10.9 9.9
ACTH, pg/ml 15 15 17 19
Subject 3
Scr, mg/dl 2.2 2.1 1.9 1.9 1.8 1.8 1.9
Salb, g/dl 3.1 3.1 3.0 3.2 3.2 3.5 3.7
UPCR, g/g 3.80 3.59 3.36 3.28 4.00 3.56 3.88
Cholesterol, mg/dl 332 345 309 341 330 353 241
Cortisol, g/dl 16.7 9.3 9.9 11.0 7.6 11.6 15.5
ACTH, pg/ml 37 22 16 23 28 29 31
Subject 4
Scr, mg/dl 1.7 1.6 1.3 1.3 1.3 1.2 1.2
Salb, g/dl 2.9 3.0 3.7 3.5 4.0 3.9 4.0
UPCR, g/g 2.23 0.62 1.05 1.37 0.71 0.25 0.84
Cholesterol, mg/dl 243 241 261 232 229 213 236
Cortisol, g/dl 9.5 10.4 9.9 8.3 7.5 11.7 6.8
ACTH, pg/ml 4 11 8 10 8 7 8
Subject 5
Scr, mg/dl 2.9 2.1 1.8 1.4 1.5 1.4 1.4
Salb, g/dl 2.1 2.1 2.5 2.9 3.5 3.2 3.4
UPCR, g/g 12.46 12.60 6.29 7.21 4.92 4.06 3.14
Cholesterol, mg/dl 201 216 218 225 158 146 157
Cortisol, g/dl 12.6 9.8 11.3 8.7 11.4 8.4 6.8
ACTH, pg/ml 14 15 19 16 14 10 12
Cholesterol = Total cholesterol; Scr = serum creatinine; Salb = serum albumin.
ACTH for Resistant Glomerular Diseases Am J Nephrol 2012;36:58–67
63
g/g) at 7 months’ follow-up since discontinuing ACTH.
As a group, the 5 subjects with IgA nephropathy went
from a pre-ACTH median (range) proteinuria of 1.59
mg/g (0.61–4.12) to a post-ACTH median (range) pro-
teinuria of 0.85 mg/g (0.21–3.51).
An independent effect of ACTH on total cholesterol
and LDL levels was not apparent. Improvements in these
lipid parameters mirrored improvements in proteinuria
in those subjects who demonstrated a response to therapy
but were unchanged for those subjects who did not re-
spond ( tables 2–4 ). No significant changes in serum cor-
tisol or ACTH levels, drawn immediately prior to ACTH
injections, were detected in any subject during the treat-
ment period.
Three subjects discontinued therapy early due to ad-
verse events. Subjects 1 and 2, both with MN and previ-
ous diagnoses of diet-controlled diabetes, had worsened
glycemic control prompting initiation of oral hypoglyce-
mic therapy. Subject 1 withdrew at 12 weeks, and subject
2 withdrew at 16 weeks. Neither had demonstrated any
Table 3. Detailed results from 5 subjects with resistant MCD or FSGS
Baseline Week 4 Week 8 Week 12 Week 16 Week 20 Week 24
Subject 6
Scr, mg/dl 0.7 0.6 0.6 0.6 0.5 0.6 0.6
Salb, g/dl 2.5 2.6 2.5 2.7 2.8 2.4 2.4
UPCR, g/g 4.76 2.29 2.13 3.32 3.28 5.30 11.35
Cholesterol, mg/dl 453 384 432 443 527 332*
Cortisol, g/dl 19.2 7.3 3.5 7.4 5.5 5.4 5.7
ACTH, pg/ml 47 14 13 12 10 13 12
Subject 7
Scr, mg/dl 0.6 0.6 0.7 0.6 0.6 0.6 0.5
Salb, g/dl 3.4 3.3 4.0 3.6 3.7 3.8 3.7
UPCR, g/g 3.16 1.69 1.22 2.64 1.21 1.51 0.78
Cholesterol, mg/dl 289 234 244 210 228 219 210
Cortisol, g/dl 20.0 10.4 11.3 13.0 7.6 7.7 11.6
ACTH, pg/ml 15 9 6 9 7 10 6
Subject 8
Scr, mg/dl 1.2 1.2 1.3 1.2 1.3 1.5 1.4
Salb, g/dl 3.7 3.8 3.4 3.2 3.8 4.0 3.9
UPCR, g/g 1.94 1.68 1.98 3.27 0.51 0.30 0.43
Cholesterol, mg/dl 223 188 166 166 168 163 165
Cortisol, g/dl 5.5 6.1 7.6 5.5 6.5 8.4 9.8
ACTH, pg/ml 7 9 8 15 8 9
Subject 9
Scr, mg/dl 0.8 0.7 0.8 0.8 1.0 0.9 1.0
Salb, g/dl 2.8 2.7 2.7 2.6 2.4 2.4 2.3
UPCR, g/g 1.96 2.58 4.00 3.12 2.43 3.85 4.08
Cholesterol, mg/dl 166 183 193 167 216 231
Cortisol, g/dl 11.8 10.4 9.4 7.6 13.8 9.8
ACTH, pg/ml 32 34 32 37 36 29 34
Subject 10
Scr, mg/dl 1.0 0.9 1.0 1.2 0.9 0.9 1.0
Salb, g/dl 3.6 3.4 3.4 3.8 3.7 3.6 3.8
UPCR, g/g 1.65 2.19 1.65 2.62 1.43 1.72 1.93
Cholesterol, mg/dl 174 156 148 181 155 190
Cortisol, g/dl 16.4 10.3 8.0 12.8 12.0 19.0 17.5
ACTH, pg/ml 11 9 11 9 11 9 17
Cholesterol = Total cholesterol; Scr = serum creatinine; Salb = serum albumin.
* Subject 6 was started on atorvastatin after the week-16 visit.
Bomback /Canetta /BeckJr. /Aya lon /
Radhakrishnan
/Ap pe l
Am J Nephrol 2012;36:58–67
64
evidence of response to ACTH therapy by conventional
laboratory parameters at the time of withdrawal (al-
though subject 2’s serum showed disappearance of anti-
bodies to the PLA
2
R over the 16 weeks of treatment). Both
su bject s rema in dep end ent on o ral hy po glycemic t herapy
1 1 year from their last dose of ACTH. Subject 14 com-
plained of weight gain and Cushingoid facies by week 8,
accompanied by an average increased in blood pressure
of 20/10 mm Hg during this time. He had experienced
similar side effects with prednisone in the past. At week
12, with evidence of worsening kidney function, he was
withdrawn from the study. Four other subjects com-
plained of insomnia during the first month of therapy,
typically on the night of injections; these symptoms abat-
ed by month 2 for these subjects. Subjects 4 and 10 re-
ported increased pigmentation of their skin, which re-
solved within 4 weeks of discontinuing the drug. No sub-
ject developed a significant infection during the study
period.
Table 4. Detailed results from 5 subjects with resistant IgA nephropathy
Baseline Week 4 Week 8 Week 12 Week 16 Week 20 Week 24
Subject 11
Scr, mg/dl 1.7 1.7 1.8 1.9 2.0 1.8 1.7
Salb, g/dl 3.7 4.0 4.1 3.7 4.0 4.1 4.4
UPCR, g/g 1.95 1.76 1.45 1.54 1.57 1.38 1.22
Cholesterol, mg/dl 175 168 175 190 191 222 194
Cortisol, g/dl 11.7 4.6 6.7 8.7 5.3 5.0 15.0
ACTH, pg/ml 11.0 9.0 12.0 14.0 13.0 10.0 20.0
Subject 12
Scr, mg/dl 2.4 2.4 2.2 2.5 2.2 2.0 2.1
Salb, g/dl 4.0 4.0 3.7 4.3 4.3 3.9 4.1
UPCR, g/g 1.59 1.50 1.08 0.84 0.94 0.79 0.57
Cholesterol, mg/dl 171 164 156 174 165 166 154
Cortisol, g/dl 12.6 11.4 8.9 10.3 10.3 9.1
ACTH, pg/ml 14 23 14 17 16 26
Subject 13
Scr, mg/dl 0.8 0.8 0.8 0.8 1.0 0.8 0.8
Salb, g/dl 4.1 4.1 4.2 4.1 4.6 4.3 4.5
UPCR, g/g 0.61 1.16 0.40 0.29 0.14 0.14 0.21
Cholesterol, mg/dl 153 187 201 175 198 162 188
Cortisol, g/dl 16.7 15.3 19.9 10.3 20.3 17.3 11.2
ACTH, pg/ml 12 8 15 15 11 17 11
Subject 14
Scr, mg/dl 2.7 2.8 3.1 3.4
Salb, g/dl 4.0 3.8 3.8 3.4
UPCR, g/g 4.12 5.34 5.21 3.51
Cholesterol, mg/dl 215 191 209 209
Cortisol, g/dl 8.5 7.1 8.3
ACTH, pg/ml 13 12 17
Subject 15
Scr, mg/dl 1.0 1.1 1.0 1.1 1.1 1.1 1.2
Salb, g/dl 4.3 4.4 4.4 4.4 4.6 4.4
UPCR, g/g 1.51 1.74 1.32 0.84 1.03 0.85 0.85
Cholesterol, mg/dl 192 216 242 240 220 185 183
Cortisol, g/dl 16.3 14.0 10.4 16.5 13.2 8.0 12.5
ACTH, pg/ml 15 17 17 20 20 22 30
Cholesterol = Total cholesterol; Scr = serum creatinine; Salb = serum albumin.
ACTH for Resistant Glomerular Diseases Am J Nephrol 2012;36:58–67
65
Discussion
We present here the first prospective study data using
ACTH for resistant glomerular diseases. In this open-la-
bel, efficacy and safety study, we treated 15 subjects with
resistant MN, MCD, FSGS, or IgA nephropathy with
ACTH gel, 80 units twice weekly, for 6 months. Two of 5
resistant MN subjects, 2 of 5 resistant MCD/FSGS sub-
jects, and 3 of 5 resistant IgA nephropathy subjects
showed significant reductions i n proteinuria to remission
levels during the treatment period, and all but 1 subject
have sustained these remissions up to 1 year after discon-
tinuing therapy. Three subjects discontinued therapy
early due to Cush ingoid side effects, including exacerba-
tion of diabetes and weight gain. These results support
the use of ACTH gel therapy for some patients with resis-
ta nt glomeru lar disease s and, nota bly, show that an ef fect
of therapy can be seen in patients with reduced kidney
function and/or prolonged course of disease.
The data from this prospective trial add to a growing
body of literature on using ACTH for nephrotic syn-
drome that, to date, is based almost entirely on observa-
tional, retrospective case series
[6, 7, 9] . Only one previ-
ous report, from Ponticelli et al.
[11] , provides data from
a prospective study of ACTH, in which 16 subjects with
MN treated with a synthetic analogue of ACTH demon-
strated similar remission rates (14 of 16 remissions, or
87.5%) as 16 subjects with MN treated with alternating
months of steroids and cyclophosphamide (12 of 16 re-
missions, or 75%). In this study, however, ACTH was used
as a first-line immunomodulatory therapy and thus
would be expected to have a better response rate than
would be seen if used in resistant patients. A case series
published by our group reported 21 cases of idiopathic
nephrotic syndrome treated with ACTH gel outside of
research settings (i.e. by prescription), of which 18 were
considered to have resistant nephrotic syndrome, having
failed a mean 2.3 immunosuppressive regimens prior to
ACTH gel therapy (9 patients had failed at least 3 prior
therapies). Overall, 11 of 21 patients (52%) achieved a
complete or partial remission, with 4 (19%) in complete
remission. Of the 11 patients who achieved complete or
partial remission, 9 had idiopathic MN, 1 had FSGS, and
1 had IgA nephropathy
[9] . The response rates presented
here of 40–60%, depending on diagnosis, appear to con-
firm the signal seen in this previous, retrospective series.
The literature to date on the benefits of ACTH for ne-
phrotic syndrome has also been almost exclusively based
on patients with MN. Our study, therefore, provides im-
por ta nt da ta on us in g AC TH in p at ien ts wi th MCD, FS GS ,
and IgA nephropathy. Indeed, prior to this current study,
the results of ACTH treatment have been reported for
only 2 patients with FSGS
[7, 9] , 3 patients with MCD [7,
9]
, and 1 patient with IgA nephropathy [9] . We chose to
combine MCD and FSGS into one treatment group. Al-
though the courses of MCD and FSGS can be quite differ-
ent on initial presentation, particularly regarding re-
sponse to immunosuppressive therapy, treatment pat-
terns converge in the setting of steroid resistance, which
(along with failure of a second-line therapy) was a prereq-
uisite for enrollment in this study. We defined resistant
IgA nephropathy as proteinuria 1 1 g/day despite effective
inhibition of the RAAS; this proteinuria cutoff has been
shown to be a strong predictor of progression to renal fail-
ure
[13 ] and previous studies have shown that use of R AAS
blockade to achieve goal proteinuria ! 1 g/day leads to sig-
nificantly better outcomes
[14 ] . Our patients, thus, have
essentially failed the standard-of-care for IgA nephropa-
thy and are at increased risk for disease progression, as
demonstrated by signficantly reduced eGFR in 3 of the 5
subjects. The benefits of ACTH therapy in this IgA popu-
lation should be compared not against RAAS blockade
therapy, which has been studied in nonresistant popula-
tions
[14 ] , but rather against steroid therapy, which has
been studied in comparable patient populations
[15 , 16] .
These resu lts should be interpreted in light of the study
population of resistant patients. In all of the glomerular
diseases included in this study, failure of first-line ther-
apy increases the probability for failure of second-line
therapy, failure of second-line therapy increases the prob-
ability for failure of third-line therapy, and so forth. The
response rates of 2/5 or 3/5 patients are impressive for
such a population and place ACTH alongside other third-
or fourth-line therapies, such as rituximab and MMF, in
terms of expected benefits and strength of supportive
data
[17 –2 2] . In addition, by restricting the use of ACTH
in this study to patients with previous treatment resis-
tance, we have reduced the possibility of spontaneous re-
mission of disease confounding interpretation of clinical
improvements, which can happen in up to one-third of
patients with MN and a much smaller percentage of pa-
tients with IgA nephropathy. Our study also gives some
guidance on when and how the drug should be used, sug-
gesting that prolonged duration of disease and moderate
renal dysfunction are not contraindications, and that
treatment responses may take up to 3 or 4 months to
manifest. The small sample presented here does not pro-
vide any evidence supporting the use of ACTH for calci-
neurin inhibitor-dependent patients, unlike previous re-
ports using rituximab
[23] .
Bomback /Canetta /BeckJr. /Aya lon /
Radhakrishnan
/Ap pe l
Am J Nephrol 2012;36:58–67
66
The data from this study are limited, however, by the
small subject sample, the lack of a control group, and the
relatively short-term follow-up. This open-label pilot
study was not intended and clearly not powered to test
hypotheses. These data do demonstrate treatment re-
sponses as well as provide safety profiles for patients with
glomerular diseases who could be candidates for ACTH
therapy. These data should be viewed, in our opinion, as
stronger than retrospective and observational data previ-
ously reported by our group and others
[6–10] , but not as
robust as those provided by Ponticelli et al. in their ran-
domized trial of synthetic ACTH
[11] nor by those data
that will emerge from the ongoing randomized trial
of ACTH gel in resistant MN (NCT01386554). Those sub-
jects who began treatment with low urine protein:crea -
tinine ratio values (i.e. ! 2.0 g/g) conceivably may have
already achieved a partial remission of disease prior to
ACTH therapy, which would increase their likelihood of
showing a response to ACTH. Finally, the relatively short
duration of follow-up in these subjects does not provide
sufficient data on the sustainability of the responses doc-
umented here. In previous reports with synthetic ACTH
from Europe, a number of subjects who have responded
to therapy have required one or more additional courses
of ACTH for relapsing disease before achieving a sus-
tained remission
[7] .
In conclusion, we present here the first prospective
data on using ACTH as therapy for resistant glomerular
diseases. In a small sample of patients with MN, FSGS,
MCD, and IgA nephropathy who had not previously re-
sponded to conventional therapies, ACTH yielded a 40–
60% response rate depending on diagnosis. For all but
one subject, remission of proteinuria was sustained up to
1 year from cessation of therapy. Despite a 20% rate of
Cushingoid side effects, the drug was otherwise well tol-
erated with no significant infections. We believe that
these data support the further investigation of ACTH as
third- or fourth-line immunosuppressive therapy for pa-
tients with resistant glomerular diseases.
Acknowledgements
We are grateful to Angela Cha, Irma Orbe, and Melanie Foley
for their assistance with conduct of this study at the Clinical Re-
se ar ch O ff ic es o f the D iv isi on o f Ne phr olo gy at C olu mbia U niv er-
sity. These data were originally presented in abstract form at the
2011 American Society of Nephrology Renal Week.
Disclosure Statement
This study was supported by an investigator initiated study
grant from Questcor Pharmaceuticals, the manufacturer of
ACTH gel, and by the Center for Glomerular Diseases at Colum-
bia University. The investigators were responsible for study de-
sign, implementation, data analysis, and manuscript preparation.
Drs. Bomback, Beck, and Appel have received consulting hono-
raria from Questcor Pharmaceuticals. Drs. Canetta, Ayalon, and
Radhakrishnan have no relevant disclosures.
References
1 Waldman M, Crew RJ, Valeri A, Busch J,
Stokes B, Markowitz G, D’Agati V, Appel G:
Adult minimal-change disease: clinical
characteristics, treatment, and outcomes.
Clin J Am Soc Nephrol 2007;
2: 445–453.
2 Cattr an DC, Appel GB, Heber t LA, Hunsick-
er LG, Pohl MA, Hoy WE, Max well DR, Ku-
nis CL: Cyclosporine in patients with ste-
roid-resistant membranous nephropathy: a
randomized trial. Kidney Int 2001;
59: 1484–
149 0.
3 Perna A, Schieppati A, Zamora J, Giuliano
GA, Braun N, Remuzzi G: Immunosuppres-
sive treatment for idiopathic membranous
nephropathy: a systematic review. Am J Kid-
ney Dis 2004;
44: 385–401.
4 Cattr an DC, Appel GB, Heber t LA, Hunsick-
er LG, Pohl MA, Hoy WE, Max well DR, Ku-
nis CL: A ra ndomized tr ial of cyclospor ine in
patients with steroid-resistant focal segmen-
tal glomerulosclerosis. North America Ne-
phrotic Syndrome Study Group. Kidney Int
1999;
56: 2220–2226.
5 Troyanov S, Wall CA, Miller JA, Scholey JW,
Cattran DC: Focal and segmental glomeru-
losclerosis: def inition and releva nce of a par-
tial remission. J Am Soc Nephrol 2005;
16:
1061–1068.
6 Rauen T, Michaelis A, Floege J, Mertens PR:
Case series of idiopathic membranous ne-
phropathy with long-term beneficial effects
of ACTH peptide 1–24. Clinical Neph rology
2009;
71: 637–642.
7 Berg AL, Arnadottir M: ACTH-induced im-
provement in the nephrotic syndrome in pa-
tients wit h a variety of diagnoses. Nephrol
Dial Transplant 2004;
19: 1305–1307.
8 B erg AL , N il ss on- Eh le P, Ar na do tt ir M: B en -
eficia l effects of ACTH on the serum lipo-
protein profile and glomerular function in
patients with membranous nephropathy.
Kidney Int 1999;
56: 1534–1543.
9 Bomback AS, Tumlin JA, Baranski J, Bour-
deau JE, Besarab A, Appel AS, Radhakrish-
nan J, Appel GB: Treatment of ne phrotic syn-
drome with adrenocorticotropic hormone
(ACTH) gel. Drug Design Dev Ther 2011;
5:
147–153.
10 Lindskog A, Ebefors K, Johansson ME, Ste-
fansson B, Granqvist A, Arnadottir M, Berg
AL, Nystrom J, Haraldsson B: Melanocortin
1 receptor agonists reduce proteinuria. J Am
Soc Nephrol 2010;
21: 1290–1298.
ACTH for Resistant Glomerular Diseases Am J Nephrol 2012;36:58–67
67
11 Ponticelli C, Passerini P, Salvadori M, Man-
no C , Vi ola B F, Pas qua li S , Mandol fo S, Mes -
sa P: A randomized pilot trial comparing
methylprednisolone plus a cytotoxic agent
versus synthetic adrenocorticotropic hor-
mone in idiopathic membranous nephropa-
thy. Am J Kidney Dis 2006;
47: 233–240.
12 Beck LH Jr, Bonegio RG, Lambeau G, Beck
DM, Powell DW, Cummins TD, Klein JB,
Salant DJ: M-type phospholipase A2 recep-
tor as target antigen in idiopathic membra-
nous nephropathy. N Engl J Med 2009;
361:
11–21.
13 Reich HN, Troyanov S, Scholey JW, Cattran
DC: Remission of proteinuria improves
prognosis in IgA nephropathy. J Am Soc
Nephrol 2007;
18: 3177–3183.
14 Praga M, Gutierrez E, Gonzalez E, Morales
E, Herna ndez E: Treatment of IgA nephropa-
thy with ACE inhibitors: a randomized and
controlled trial. J Am Soc Nephrol 2003;
14:
1578–1583.
15 Manno C, Torres DD, Rossini M, Pesce F,
Schena FP: Randomized controlled clinical
trial of corticosteroids plus ACE-inhibitors
with long-term follow-up in proteinuric IgA
nephropathy. Nephrol Dia l Transpla nt 2009;
24: 3694–3701.
16 Lv J, Zhang H, Chen Y, Li G, Jiang L, Singh
AK, Wang H: Combination therapy of pred-
nisone and ACE i nhibitor versus ACE-in hib-
itor therapy alone in patients with IgA ne-
phropathy: a randomized controlled trial.
Am J Kidney Dis 2009;
53: 26–32.
17 Bomback AS, Derebail VK, McGregor JG,
Kshir sagar AV, Fa lk RJ, Nachma n PH: Ritux-
imab ther apy for membranous nephropathy :
a systematic review. Clin J Am Soc Nephrol
2009;
4: 734–744.
18 Fervenza FC, Cosio FG, Erickson SB, Specks
U, Herzenberg AM, Dillon JJ, Leung N, Co-
hen IM, Wochos DN, Bergst ralh E, H ladune-
wich M, Cattran DC: Rituximab treatment
of idiopathic membranous nephropathy.
Kidney Int 2008;
73: 117–125.
19 Dussol B, Morange S, Burtey S, Indreies M,
Cassuto E, Mourad G, Villar E, Pouteil-No-
ble C, Kar aaslan H, Sichez H, L asseur C, Del-
mas Y, Nogier MB, Fat hallah M, L oundou A,
Mayor V, Berland Y: Mycophenolate mofetil
monotherapy in membranous nephropathy:
a 1-year randomized controlled trial. Am J
Kidney Dis 2008;
52: 699–705.
20 Palmer SC, Nand K, Strippoli GF: Interven-
tions for minimal change disease in adults
with nephrotic syndrome. Cochrane Data-
base System Rev (online) 2008:CD001537.
21 Cattran DC, Wang MM, Appel G, Matalon
A, Briggs W: Mycophenolate mofetil in the
treatment of focal segmenta l glomeruloscle-
rosis. Clin Nephrol 2004;
62: 405–411.
2 2 Fe rna nde z-Fre sne do G, Seg arr a A, G onz ale z
E, Alexandru S, Delgado R, Ramos N, Egido
J, Praga M: R ituximab t reatment of adult pa-
tients with steroid-resistant focal segmenta l
glomerulosclerosis. Clin J Am Soc Nephrol
2009;
4: 1317–1323.
23 Segarra A, Praga M, Ramos N, Polanco N,
Cargol I, Gutierrez-Solis E, Gomez MR,
Montoro B, Camps J: Successful treatment
of membranous glomerulonephritis with
rituximab in calcineurin inhibitor-depen-
dent patients. C lin J Am Soc Nephrol 2 009;
4:
1083–1088.
... Clinical studies (randomized controlled studies, open-label studies), retrospective case series, and case reports have shown that RCI may be effective in achieving complete or partial remission of proteinuria in NS. [74][75][76][77][78][79] For example, in a combined prospective trial and retrospective review of 24 patients with proteinuria due to FSGS, 7 patients achieved remission with RCI. 74 Similarly, a prospective open-label trial of RCI showed that 8 of 15 patients with various NS etiologies saw a reduction in proteinuria; this included 2 of 5 patients with resistant idiopathic MN, 2 of 5 patients with MCD or FSGS, and 4 of 5 patients with IgAN. ...
... No patient developed a significant infection during the study. 75 ...
Article
Full-text available
Acthar® Gel (repository corticotropin injection [RCI]) is a naturally sourced complex mixture of adrenocorticotropic hormone analogs and other pituitary peptides used to treat patients with serious and rare inflammatory and autoimmune conditions. This narrative review summarizes the key clinical and economic findings among 9 indications: infantile spasms (IS), multiple sclerosis (MS) relapses, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), dermatomyositis and polymyositis (DM/PM), ocular inflammatory diseases (primarily uveitis and severe keratitis), symptomatic sarcoidosis, and proteinuria in nephrotic syndrome (NS). Key studies of clinical efficacy and healthcare resource utilization and cost from 1956 to 2022 are discussed. Evidence supports the efficacy of RCI across all 9 indications. RCI is recommended as first-line treatment for IS and is associated with improved outcomes for the other 8 indications, including increased recovery rates in MS relapse; improved disease control in RA, SLE, and DM/PM; real-world effectiveness in patients with uveitis and severe keratitis; improved lung function and reduced corticosteroid use in symptomatic sarcoidosis; and increased rates of partial remission of proteinuria in NS. For many indications, RCI may improve clinical outcomes during exacerbations or when conventional treatments have failed to show a benefit. RCI is also associated with a reduction in the use of biologics, corticosteroids, and disease-modifying antirheumatic drugs. Economic data suggest RCI is a cost-effective, value-based treatment option for MS relapse, RA, and SLE. Other economic benefits have been demonstrated for IS, MS relapses, RA, SLE, and DM/PM, including reduced hospitalizations, lengths of stay, inpatient and outpatient services, and emergency department visits. RCI is considered safe and effective and features economic benefits for numerous indications. Its ability to control relapse and disease activity makes RCI an important nonsteroid treatment option that could help preserve functioning and well-being among patients with inflammatory and autoimmune conditions.
... Immunosuppressants such as adrenocorticotropic hormone (ACTH) gel and similar analogues have been used with moderate success in treating some GN cases, particularly in patients with membranous nephropathy. 27,28 Investigators seeking to evaluate its utility in treating FSGS have reported complete or partial remission in approximately 30% of patients treated with biweekly subcutaneous ACTH injections. 27 Furthermore, multiple studies of ACTH for steroid-unresponsive idiopathic FSGS patients have been plagued by high attrition and a significant incidence of adverse effects as a result of treatment. ...
... 27 Furthermore, multiple studies of ACTH for steroid-unresponsive idiopathic FSGS patients have been plagued by high attrition and a significant incidence of adverse effects as a result of treatment. 27,28 Despite trends towards remission of proteinuria demonstrated in these studies, the lack of robust benefit and frequency of adverse effects associated with ACTH therapy bars it from consideration as a plausible second-line treatment currently. Furthermore, ACTH therapy is extremely expensive, which creates a financial barrier to using it in exploratory or clinical settings. ...
Article
Purpose: Idiopathic focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD) are chronic glomerulopathies which may compromise patients’ quality of life, and for which there is no cure. This literature review aimed to summarize our current understanding of the pathophysiology, clinical characteristics, and best available treatment for the two conditions in order to outline a consolidated treatment protocol and identify future research considerations. Methods: PubMed was systematically searched by a single reviewer in order to identify primary studies pertaining to the diagnosis, treatment and classification of FSGS and MCD. Additionally, a hand search of UpToDate was conducted to glean further information about the best available evidence as summarized for clinician use. Relevant information was extracted and synthesized. Results: Primary FSGS and MCD result from distinct pathogenic mechanisms, hypothesized to involve kidney injury via immune dysregulation. Patients require a kidney biopsy for diagnostic purposes. First-line treatment involves glucocorticoids (i.e. prednisone), although patients’ responsiveness may be inconsistent; second-line treatment is immunotherapy. Conclusion: This review summarized clinically-important information about FSGS and MCD, and emphasized the need for further research in the field of clinical nephrology. Large scale trials such as the Cure Glomerulonephropathy should be conducted to gather information about the affected population.
... 9,10 Furthermore, a number of uncontrolled case series show that ACTH monotherapy is still effective in pMN patients who are resistant to steroid-containing regimens. 6,11,12 In addition to being a core component of the hypothalamic-pituitary-adrenal axis, ACTH is also a key member of the melanocortin hormone system, which consists of 5 melanocortin receptors (MC1~5R) and multiple endogenous melanocortin ligands. 4 Burgeoning evidence suggests that the melanocortinergic signaling pathway protects against glomerular diseases, and may be responsible for the beneficial effects of ACTH in MN. 6 However, the molecular mechanism of this renoprotective activity remains elusive. ...
... 4 Burgeoning evidence suggests that the melanocortinergic signaling pathway protects against glomerular diseases, and may be responsible for the beneficial effects of ACTH in MN. 6 However, the molecular mechanism of this renoprotective activity remains elusive. Of note, the beneficial effect of ACTH in pMN patients, even in those resistant to steroids, had been preceded by receding levels of anti-PLA2R autoantibodies, 10,12 suggesting that melanocortins may modulate the humoral immunity. To test this hypothesis, this study employed the rat model of passive Heymann nephritis (PHN), a model of MN, 1,13 and tested the efficacy of diverse melanocortins and analogues, including Repository Corticotropin Injection (RCI) containing the natural ACTH 1-39 , 6 and the non-steroidogenic pan-MCR agonist [Nle 4 , DPhe 7 ]-α-MSH (NDP-MSH) 6 that has been recently approved to treat porphyria. ...
Article
Background: The pituitary neuropeptide melanocortins, and specifically ACTH, have recently emerged as a novel therapeutic modality for membranous nephropathy (MN). However, the mechanism(s) of action remains elusive. Methods: Passive Heymann nephritis (PHN), a model of MN, was induced in wild-type (WT) rats and melanocortin 1 receptor (MC1R) knockout (KO) rats generated by the CRISPR/Cas9 technology, followed by treatment with various melanocortin agents, including Repository Corticotropin Injection, the non-steroidogenic pan-MCR agonist NDP-MSH, and the selective MC1R agonist MS05. Additional rats received adoptive transfer of syngeneic bone marrow-derived cells (BMDC) beforehand. Kidney function, histology and molecular changes were evaluated. Results: MC1R KO worsened PHN, associated with increased deposition of autologous IgG and complement C5b-9 in glomeruli and higher circulating levels of autologous IgG, as evidence of a sensitized humoral immune response. Melanocortin therapy ameliorated PHN in WT rats, coinciding with reduced glomerular deposition of autologous IgG and C5b-9. The beneficial efficacy of melanocortins was blunted in KO rats but was restored by adoptive transfer of syngeneic BMDC derived from WT rats. Mechanistically, MC1R was expressed in B lymphocytes, and negatively associated with B cell activation as revealed by gene set enrichment analysis. MC1R agonism triggered MITF induction in activated B cells in a cAMP-dependent mode, and repressed the expression of IRF4, a lymphoid transcription factor essential for B cell development and maturation, resulting in suppressed plasmacytic differentiation and IgG production. Conclusion: MC1R signaling negatively modulates B cell activation and suppresses humoral immune responses in PHN, representing a novel therapeutic target for MN.
... Bortezomib reduced proteinuria in IgAN patients, but the study was non-randomized, had a small sample size, and lacked a control group [14]. Additionally, proteinuria remissions were also achieved in some patients with refractory nephropathy treated with ACTH Gel [15]. ...
... And several common traits were shared in these trials, such as low enrollment and short follow-up time. Notably, these trials did provide new ideas for treating IgAN, such as the effect of Bortezomib on improving renal function and urinary protein of IgAN patients [14], and the application of ACTH Gel in refractory IgAN [15]. Nevertheless, both trials had small sample sizes and no control, thus the exact efficacy needs further validation. ...
Article
Full-text available
Objectives IgA Nephropathy (IgAN) is common chronic kidney disease with a high incidence. This study aims to analyze comprehensively therapeutic clinical trials for IgAN registered on ClinicalTrials.gov. Methods Therapeutic trials for IgAN registered on ClinicalTrials.gov. up to 15 August 2021 were obtained. The general characteristics, features of experimental design, treatment strategies, and some main inclusion criteria and outcome measures were accessed. Results A total of 104 therapeutic clinical trials for IgAN were extracted on ClinicalTrials.gov up to 15 August 2021. Most of these trials explored the treatment for primary IgAN confirmed by renal biopsy in adults. Only 9% of all selected trials had results. Forty-five percent of trials recruited 50 or fewer participants, and 73% were adults or older adults. 99% of trials were interventional studies, and of all the interventional trials, 70% of trials were randomized, and 68% exercised a parallel assignment of intervention model. Immunosuppression was the most studied for the treatment of IgAN. Moreover, many novel agents had been increasingly studied in recent years. Furthermore, the inclusion criteria and primary outcome measures in these trials were diverse, and the level of proteinuria and change of proteinuria levels were the most used as inclusion criteria and primary outcome, respectively. Conclusions The majority of therapeutic trials for IgAN were randomized, none masking and parallel-assignment interventional studies, primarily recruiting adult patients as research subjects. These trials had relatively small sample sizes and short observation. Thus, more large-scale, multicenter, and randomized controlled trials are still needed to improve the management for IgAN.
... A small observational study tested the effects of adrenocorticotropic hormone (ACTH) gel (80 units subcutaneously twice weekly) in 15 subjects with resistant glomerular diseases (MCD = 2, FSGS = 3) for 6 months. One subject with resistant FSGS achieved CR and one subject with resistant MCD achieved PR but relapsed within 4 weeks of stopping ACTH [116]. The evidence should be studied further in controlled trials against currently available therapies for resistant disease. ...
Chapter
Full-text available
Minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) are the two key forms of idiopathic nephrotic syndrome. This chapter examines the evidence for management of MCD and FSGS in adults. Minimal change disease presents as nephrotic syndrome of sudden onset. In adults proteinuria is less selective than in children. MCD and FSGS are both examples of pathogenic mechanisms that primarily affect the podocyte. Abnormal T cell dysfunction has long been implicated. However, no consensus has yet been reached regarding a single cell subset. Response to steroid therapy was the most important factor for preservation of renal function. Infectious complications are commonly seen in nephrotic patients due to mass proteinuria and use of immunosuppressive agents. Corticosteroids remain the mainstay of first‐line treatment of MCD in adults. Current consensus remains that corticosteroids with full dose and extended period of time are the first‐line of therapy in nephrotic FSGS.
... Three patients had previously received CNIs, two cyclophosphamide, two steroids, four MMF, and one patient previously received rituximab. Two of the five patients achieved partial clinical remission on ACTH therapy and three patients achieved immunological remission (92). ...
Article
Full-text available
Primary membranous nephropathy (pMN) is an auto-immune disease characterized by auto-antibodies targeting podocyte antigens resulting in activation of complement and damage to the glomerular basement membrane. pMN is the most common cause of nephrotic syndrome in adults without diabetes. Despite a very heterogeneous course of the disease, the treatment of pMN has for many years been based on uniform management of all patients regardless of the severity of the disease. The identification of prognostic markers has radically changed the vision of pMN and allowed KDIGO guidelines to evolve in 2021 towards a more personalized management based on the assessment of the risk of progressive loss of kidney function. The recognition of pMN as an antibody-mediated autoimmune disease has rationalized the use immunosuppressive drugs such as rituximab. Rituximab is now a first line immunosuppressive therapy for patients with pMN with proven safety and efficacy achieving remission in 60-80% of patients. For the remaining 20-40% of patients, several mechanisms may explain rituximab resistance: (i) decreased rituximab bioavailability; (ii) immunization against rituximab; and (iii) chronic glomerular damage. The treatment of patients with rituximab-refractory pMN remains controversial and challenging. In this review, we provide an overview of recent advances in the management of pMN (according to the KDIGO 2021 guidelines), in the understanding of the pathophysiology of rituximab resistance, and in the management of rituximab-refractory pMN. We propose a treatment decision aid based on immunomonitoring to identify failures related to underdosing or immunization against rituximab to overcome treatment resistance.
... In the United States, the effects of a natural ACTH gel formulation were assessed in 11 patients with PMN. Two participants entered complete and seven a partial remission, while two failed to respond (61). In another study, 20 patients with MN and nephrotic syndrome received a subcutaneous dose of 40 or 80 IU ACTH twice weekly. ...
Article
Full-text available
Primary Membranous Nephropathy (PMN) is the most frequent cause of nephrotic syndrome in adults. If untreated, PMN can lead to end-stage renal disease; moreover, affected patients are at increased risk of complications typical of nephrotic syndrome such as fluid overload, deep vein thrombosis and infection. The association of PMN with HLA-DQA1 and the identification in around 70% of cases of circulating autoantibodies, mainly directed towards the phospholipase A2 receptor, supports the autoimmune nature of the disease. In patients not achieving spontaneous remission or in the ones with deteriorating kidney function and severe nephrotic syndrome, immunosuppression is required to increase the chances of achieving remission. The aim of this review is to discuss the evidence base for the different immunosuppressive regimens used for PMN in studies published so far; the manuscript also includes a section where the authors propose, based upon current evidence, their recommendations regarding immunosuppression in the disease, while highlighting the still significant knowledge gaps and uncertainties.
... 3 The majority of the literature for GN involves natural ACTH. [4][5][6][7][8] The cases series for use of synthetic ACTH in GN are small, ranging from 2 to 14 patients. 9,10 A recent systematic review suggested that ACTH may be used in a variety of glomerular diseases; however, that review consisted of cases primarily treated with the natural form. ...
Article
Full-text available
Rationale Synthetic adrenocorticotropic hormone (Tetracosactide) has been used in the treatment of refractory glomerular diseases. Literature surrounding the use of this medication is limited to small case series and there is conflicting data on the rate of adverse events associated with this medication. Presenting concerns of the patient Glomerulonephritis not in remission after at least 6 months of treatment with conservative care. Stable doses of concurrent immunosuppression were permitted. Diagnoses Membranous nephropathy, IgA nephropathy, minimal change disease, and focal and segmental glomerulosclerosis. Intervention Intramuscular synthetic adrenocorticotropic hormone (Tetracosactide, Synacthen Depot) with doses of either 1 mg weekly or 1 mg twice weekly. Outcomes Five of 12 patients had at least a partial remission with Tetracosactide. Median time to response was 6 months for responders. Five of the 12 patients had adverse events documented, 2 of which led to treatment discontinuation. No patients with focal and segmental glomerulosclerosis responded to treatment. Lessons Learned Higher rate of adverse events than previously reported with synthetic adrenocorticotropic hormone and uncertain treatment efficacy.
Article
The melanocortin hormone system has emerged as a novel therapeutic target for treating refractory glomerular diseases. However, the role of hematopoietic melanocortin 1 receptor (MC1R) signaling remains unknown. Upon insult by rabbit nephrotoxic serum, MC1R null-mutant mice developed more severe crescentic glomerulonephritis than wild-type mice, marked by aggravated proteinuria, kidney dysfunction and histologic lesions. Melanocortin therapy, using Repository Corticotropin Injection (Acthar Gel), the pan-melanocortin receptor agonist NDP-MSH, or the MC1R agonist MS05, ameliorated experimental nephritis in wild-type mice but this effect was blunted in null mice. Exacerbated experimental nephritis in null mice was associated with increased glomerular deposition of autologous IgG and C5b-9, in parallel with higher circulating levels of autologous IgG2c and IgG3. Additionally, the Th1 immune response was potentiated in null mice with experimental nephritis, accompanied by diminished kidney FoxP3⁺ regulatory T cells. Kidney infiltration of macrophages was also augmented by MC1R deficiency with an enhanced M1 polarization. Moreover, adoptive transfer of syngeneic bone marrow-derived cells from wild-type mice mitigated experimental nephritis in null mice and restored the beneficial efficacy of melanocortins. Mechanistically, MC1R was expressed by diverse subsets of kidney leukocytes, including macrophages, T and B lymphocytes, and inversely associated with the NFκB pathway, a key player in immune responses. MS05 attenuated the production of rabbit IgG-specific IgG2c and IgG3 in cultured wild-type splenocytes, and promoted M2 polarization in M1-primed wild-type macrophages, associated with NFκB inhibition. In contrast in null splenocytes or macrophages, this effect of MS05 was barely detectable, but was mimicked by an NFκB inhibitor. Thus, hematopoietic MC1R signaling attenuates experimental nephritis and mediates the beneficial effect of melanocortin therapy via, in part, by regulating the immune response.
Article
Full-text available
A synthetic adrenocorticotropin (ACTH) analog has shown efficacy in Europe as primary and secondary therapy for nephrotic syndrome, but there is no published experience using the natural, highly purified ACTH gel formulation, available in the United States, for nephrotic syndrome. We therefore investigated the use of ACTH gel for nephrotic syndrome in the United States. Twenty-one patients with nephrotic syndrome treated with ACTH gel outside of research settings in the United States, with initiation of therapy by December 31, 2009, allowing a minimum 6 months follow-up. We defined complete remission as stable renal function with proteinuria falling to <500 mg/day, and partial remission as stable renal function with >50% reduction in proteinuria from 500 to 3500 mg/day. Twenty-one patients with nephrotic syndrome were treated: 11 with idiopathic membranous nephropathy (iMN), 4 with membranoproliferative glomerulonephritis (MPGN), 1 with focal segmental glomerulosclerosis (FSGS), 1 with minimal change disease (MCD), 1 with immunoglobulin A (IgA) nephropathy, 1 with class V systemic lupus erythematosus (SLE) glomerulonephritis, 1 with monoclonal diffuse proliferative glomerulonephritis, and 1 with unbiopsied nephrotic syndrome. ACTH was used as primary therapy for 3 patients; the remaining patients had previously failed a mean 2.3 immunosuppressive regimens. Eleven patients achieved a complete or partial remission, with 4 (19%) in complete remission. Of the 11 patients who achieved remission, 9 had iMN, 1 had FSGS, and 1 had IgA nephropathy. Of the 11 patients with iMN, 3 (27%) achieved complete remission and 6 (55%) achieved partial remission despite having previously failed a mean 2.4 therapies. Five patients reported steroid-like adverse effects, but there were no severe infections. The limitations were retrospective data analysis with short-term follow-up. ACTH gel may be a viable treatment option for resistant nephrotic syndrome due to membranous nephropathy. Short-term data suggest that remission rates may approach 80%.
Article
Full-text available
Membranous nephropathy is one of the most common causes of nephrotic syndrome in adults. Recent reports suggest that treatment with adrenocorticotropic hormone (ACTH) reduces proteinuria, but the mechanism of action is unknown. Here, we identified gene expression of the melanocortin receptor MC1R in podocytes, glomerular endothelial cells, mesangial cells, and tubular epithelial cells. Podocytes expressed most MC1R protein, which colocalized with synaptopodin but not with an endothelial-specific lectin. We treated rats with passive Heymann nephritis (PHN) with MS05, a specific MC1R agonist, which significantly reduced proteinuria compared with untreated PHN rats (P < 0.01). Furthermore, treatment with MC1R agonists improved podocyte morphology and reduced oxidative stress. In summary, podocytes express MC1R, and MC1R agonism reduces proteinuria, improves glomerular morphology, and reduces oxidative stress in nephrotic rats with PHN. These data may explain the proteinuria-reducing effects of ACTH observed in patients with membranous nephropathy, and MC1R agonists may provide a new therapeutic option for these patients.
Article
Full-text available
Immunoglobulin A nephropathy (IgAN) is the most common cause of chronic renal failure among primary glomerulonephritis patients. The best treatment for IgAN remains poorly defined. We planned a long-term, prospective, open-label, multicentre, centrally randomized controlled trial to assess whether the combination of prednisone and ramipril was more effective than ramipril alone in patients with proteinuric IgAN. Ninety-seven biopsy-proven IgAN patients with moderate histologic lesions, 24-h proteinuria > or =1.0 g and estimated glomerular filtration rate (eGFR) > or = 50 ml/min/ 1.73 m(2) were randomly allocated to receive a 6-month course of oral prednisone plus ramipril (combination therapy group) or ramipril alone (monotherapy group) for the total duration of follow-up. The primary outcome was the progression of renal disease defined as the combination of doubling of baseline serum creatinine or end-stage kidney disease (ESKD). The secondary outcomes were the rate of renal function decline defined as the eGFR slope over time, and the reduction of 24-h proteinuria. After a follow-up of up to 96 months, 13/49 (26.5%) patients in the monotherapy group reached the primary outcome compared with 2/48 (4.2%) in the combination therapy group. The Kaplan-Meier analysis showed a significantly higher probability of not reaching the combined outcome in the combination therapy group than in the monotherapy group (85.2% versus 52.1%; log-rank test P = 0.003). In the multivariate analysis, baseline serum creatinine and 24-h proteinuria were independent predictors of the risk of primary outcome; treatment with prednisone plus ramipril significantly reduced the risk of renal disease progression (hazard ratio 0.13; 95% confidence interval 0.03-0.61; P = 0.01). The mean rate of eGFR decline was higher in the monotherapy group than in the combination therapy group (-6.17 +/- 13.3 versus -0.56 +/- 7.62 ml/min/ 1.73 m(2)/year; P = 0.013). Moreover, the combined treatment reduced 24-h proteinuria more than ramipril alone during the first 2 years. Our results suggest that the combination of corticosteroids and ramipril may provide additional benefits compared with ramipril alone in preventing the progression of renal disease in proteinuric IgAN patients in the long-term follow-up.
Article
Full-text available
Calcineurin inhibitors (CNIs) induce remission of proteinuria in most nephrotic patients with membranous glomerulonephropathy (MGN). However, 60% of patients become treatment dependent and are at risk of chronic nephrotoxicity. The aim of this study was to evaluate the efficacy of rituximab in patients with long-term dependence on CNIs. Thirteen patients with MGN, normal renal function, and proven dependence on CNIs, despite previous treatment with other immunosuppressant drugs, received a single trial of four weekly doses of rituximab (375 mg/m(2)). Outcome measures were the percentage of patients with CNI withdrawal and no evidence of relapse and the percentage of patients with complete or partial remission 30 mo after CNI withdrawal. After rituximab, proteinuria decreased significantly (2.5 +/- 0,76 basal versus 0.85 +/- 0.17 at 6 mo; P = .0003). CNIs and other immunosuppressant drugs could be withdrawn in all patients with no evidence of relapse. After CNI withdrawal, GFR increased significantly (90.3 +/- 15 basal to 106.4 +/- 20 at 3 mo with a mean increase of 15.3% [range 0-20]). Three patients suffered a relapse of nephrotic proteinuria 19, 23, and 28 mo after rituximab treatment; all were successfully treated with a second course of rituximab. At 30 mo, all patients were in remission. In patients with MGN with long-term CNI dependence, rituximab can be an effective tool to overcome dependence on CNI, thus avoiding the risk of nephrotoxicity related to the chronic exposure to these drugs.
Article
Full-text available
In patients with idiopathic membranous nephropathy (IMN), immunosuppressive therapy is usually considered when severe nephrotic syndrome or risk for progressive renal failure exist. Recently, several studies showing beneficial effects of synthetic adrenocorticotropic hormone (ACTH) under such circumstances have been published. The objective of the present case series was to evaluate long-term ACTH effects on proteinuria and renal function. Four patients with biopsy-proven membranous nephropathy and nephrotic syndrome were enrolled (median age 50 years (range 38 - 61), median GFR 39.5 ml/min (range 20 - 62), median proteinuria 9.6 g/d (range 6.0 - 20.0). Prior immunosuppressive treatment regimens included steroids, cyclosporine A, cyclophosphamide, mycophenolate mofetil or azathioprine. The patients received a synthetic ACTH analogue intramuscularly for a median duration of 8 months (range 3 - 24). ACTH dosage was adjusted according to side effects between 0.25 and 2.25 mg/week. Follow-up lasted between 24 and 82 months after therapy initiation. All 4 patients exhibited partial (n = 2) or complete (n = 2) remissions of their nephrotic syndrome within the first year. After discontinuation of ACTH therapy, proteinuria remained low in 3 of 4 cases, whereas 1 patient exhibited undulating proteinuria. Glomerular function (as assessed by glomerular filtration rate, GFR) was maintained in all patients. Side effects were minor and included weight gain, elevated blood pressure and hyperglycemia. In all 4 cases with IMN, ACTH treatment induced a lasting disease remission with relatively few side effects.
Article
Full-text available
The treatment of membranous nephropathy (MN) remains controversial. Rituximab, which selectively targets B cells, has emerged as a possible alternative treatment option with limited toxicity. The available data on rituximab therapy for MN were reviewed using the MEDLINE database (inception to August 1, 2008), Google Scholar, and selected reference lists. English-language studies investigating the use of rituximab in idiopathic and secondary MN, in native and transplanted kidneys, were included. Study design, subject number, clinical characteristics (diagnosis, previous and concomitant treatment courses, baseline proteinuria, baseline renal function), rituximab protocol, follow-up period, achievement of complete or partial remission, changes in proteinuria and renal function, and adverse effects of therapy were extracted. Twenty-one articles were included for review; all were either case reports or case series without controls. More than half of the published cases (50 of 85) came from one center where rituximab was used as primary immunosuppression for idiopathic MN. The available data suggest that rituximab, dosed either as 375 mg/m(2) once weekly for 4 wk or as 1 g on days 1 and 15, achieves a 15 to 20% rate of complete remission and a 35 to 40% rate of partial remission. The drug was well tolerated with minimal adverse events. Although rituximab may prove to be a better treatment option for MN than alkylating agents or calcineurin inhibitors, the current literature only supports using the drug in research protocols. Whether, when, how, and why to use rituximab in MN remains to be determined.
Article
Isolated case reports have shown a beneficial effect of rituximab on pediatric patients with primary FSGS, but there is no information about rituximab treatment of FSGS in adults. All patients who had biopsy-proven FSGS and were treated with rituximab in Spain were identified, independent of their positive or negative response, among the nephrology departments that belong to the Spanish Group for the Study of Glomerular Diseases (GLOSEN). Their characteristics and outcome after rituximab treatment were studied. Eight patients were identified. Rituximab failed to improve nephrotic syndrome in five of eight patients, who continued to show massive proteinuria and exhibited a rapidly deteriorating renal function in two cases. Among the remaining three patients, two of them showed an improvement of renal function and a remarkable proteinuria reduction and one experienced a beneficial but transitory effect after rituximab. There were no differences in clinical or laboratory characteristics or in the CD20 B lymphocyte count after rituximab between these three patients and the five who had a negative response. The only difference was in the regimen of rituximab administration: Whereas the five patients with a negative response received only four weekly consecutive infusions of 375 mg/m(2), the three remaining patients received additional doses of rituximab. Only a minority (three of eight) of patients in our series of adult patients with FSGS showed a positive influence of rituximab. More studies are necessary to characterize further the optimal dosages and the mechanisms of action of rituximab in FSGS.
Article
Idiopathic membranous nephropathy, a common form of the nephrotic syndrome, is an antibody-mediated autoimmune glomerular disease. Serologic diagnosis has been elusive because the target antigen is unknown. We performed Western blotting of protein extracts from normal human glomeruli with serum samples from patients with idiopathic or secondary membranous nephropathy or other proteinuric or autoimmune diseases and from normal controls. We used mass spectrometry to analyze the reactive protein bands and confirmed the identity and location of the target antigen with a monospecific antibody. Serum samples from 26 of 37 patients (70%) with idiopathic but not secondary membranous nephropathy specifically identified a 185-kD glycoprotein in nonreduced glomerular extract. Mass spectrometry of the reactive protein band detected the M-type phospholipase A(2) receptor (PLA(2)R). Reactive serum specimens recognized recombinant PLA(2)R and bound the same 185-kD glomerular protein as did the monospecific anti-PLA(2)R antibody. Anti-PLA(2)R autoantibodies in serum samples from patients with membranous nephropathy were mainly IgG4, the predominant immunoglobulin subclass in glomerular deposits. PLA(2)R was expressed in podocytes in normal human glomeruli and colocalized with IgG4 in immune deposits in glomeruli of patients with membranous nephropathy. IgG eluted from such deposits in patients with idiopathic membranous nephropathy, but not in those with lupus membranous or IgA nephropathy, recognized PLA(2)R. A majority of patients with idiopathic membranous nephropathy have antibodies against a conformation-dependent epitope in PLA(2)R. PLA(2)R is present in normal podocytes and in immune deposits in patients with idiopathic membranous nephropathy, indicating that PLA(2)R is a major antigen in this disease.
Article
Recent studies have shown that both steroids and angiotensin-converting enzyme (ACE) inhibitors improve kidney survival and decrease proteinuria in patients with immunoglobulin A nephropathy. In this study, we aim to investigate whether the addition of steroids to ACE-inhibitor therapy produces a more potent antiproteinuric effect and better protection of kidney function than an ACE inhibitor alone. Randomized controlled trial. Patients with biopsy-proven immunoglobulin A nephropathy with proteinuria of 1 to 5 g/d of protein. 63 patients were randomly assigned to either cilazapril alone (ACE-inhibitor group; n = 30) or steroid plus cilazapril (combination group; n = 33). The primary end point was kidney survival, defined as a 50% increase in baseline serum creatinine level. After follow-up for up to 48 months, 7 patients in the ACE-inhibitor group (24.1%) reached the primary end point compared with 1 patient (3%) in the combination group. Kaplan-Meier kidney survival was significantly better in the combination group than the ACE-inhibitor group after 24 and 36 months (96.6% versus 75.7%, 96.6% versus 66.2%; P = 0.001). Urine protein excretion significantly decreased in patients in the combination group compared with the ACE-inhibitor group (time-average proteinuria, 1.04 +/- 0.54 versus 1.57 +/- 0.86 g/d of protein; P = 0.01). Multivariate analysis showed that combination treatment (hazard ratio, 0.1; 95% confidence interval, 0.014 to 0.946) and time-average proteinuria (hazard ratio, 14.3; 95% confidence interval, 2.86 to 71.92) were independent predictors of kidney survival. Small sample size, a single center, and slight imbalances at baseline. Our results suggest that the addition of steroid to ACE-inhibitor therapy provided additional benefit compared with an ACE inhibitor alone. However, this was a pilot study with a small number of participants achieving the end points, and thus further validation is necessary.
Article
Previous studies have shown that short-term treatment with adrenocorticotrophic hormone (ACTH) has a strong and rapid lipid-lowering effect. In this long-term study of nephrotic patients with idiopathic membranous nephropathy, the influence of ACTH on the serum lipoprotein profile and glomerular function as well as the dose-effect relationship was investigated. Fourteen patients received ACTH intramuscularly at increasing doses during 56 days. Serum concentrations of lipids, lipoproteins, and apolipoproteins as well as variables of glomerular function were analyzed, and the side-effects were recorded. ACTH treatment, in the estimated optimal dosage, was then continued in five patients with severe steroid-resistant nephrotic syndrome. In these five patients, the total treatment period was 12 months, and the follow-up time after discontinuing treatment was 18 months. Taking both the statistically significant therapeutic effects and the modest side-effects into consideration, the optimal dosage of ACTH was estimated to be 1 mg twice per week. At that dose, reductions by 30 to 60% in the serum concentrations of cholesterol, triglycerides, apolipoprotein B, and lipoprotein(a) were observed, whereas the serum concentrations of high-density lipoprotein cholesterol and apolipoprotein AI rose by 30 to 40%. In addition, the urinary albumin excretion decreased by 90%, and the glomerular filtration rate increased by 25%. Deterioration was observed in all cases when ACTH was discontinued after a treatment duration of 56 days. However, the five patients in whom ACTH therapy was resumed were still in remission 18 months after discontinuance of treatment. In nephrotic patients with idiopathic membranous nephropathy, treatment with ACTH 1 mg twice per week was associated with significant long-term improvements in serum lipoprotein pattern and glomerular function.