Sei sulla pagina 1di 10

In-Depth Review

Posttransplant Recurrence of Primary Glomerulonephritis


Claudio Ponticelli* and Richard J. Glassock†
*Division of Nephrology, Scientific Institute Humanitas, Milan, Italy; and †Department of Medicine, David Geffen
School of Medicine at the University of California–Los Angeles, Los Angeles, California

All forms of primary GN may recur after kidney transplantation and potentially jeopardize the survival of the graft. IgA
nephritis (IgAN) may recur in approximately one third of patients, more frequently in younger patients and in those with a
rapid progression of the original disease. However, with the exception of few patients with rapid progression, there is no
evidence that recurrence of IgAN has a deleterious effect on graft survival at least up to 10 years. Recurrence of focal segmental
glomerulosclerosis (FSGS) is often associated with nephrotic proteinuria and is more frequent in children, in patients with
rapid progression of the original disease, and in those who lost a previous transplant from recurrence. The natural course of
recurrent FSGS is usually unfavorable. Early and intensive plasmapheresis may obtain complete or partial response in several
patients. Good results have also been reported with rituximab. Idiopathic membranous nephropathy (IMN) may recur in 30%
to 40% of patients. The graft survival in patients with IMN is not different than that of patients with other renal diseases. Good
results with rituximab have been reported. Membranoproliferative GN (MPGN) may recur in 27% to 65% of patients. The
recurrence is more frequent and the prognosis is more severe in type II MPGN. Although recurrent GN is relatively frequent
and may worsen the outcome of renal allografts in some patients, its effect is diluted by several other risk-factors that may
have a greater effect than recurrent GN on the long-term graft survival.
Clin J Am Soc Nephrol 5: 2363–2372, 2010. doi: 10.2215/CJN.06720810

A
s for other patients with ESRD, renal transplantation IgA1 and IgA or IgG autoantibodies to the aberrantly glycosy-
represents the treatment of choice for patients with lated IgA1 (4,5). The disease usually runs an indolent course but
primary (idiopathic) GN and advanced, irreversible may lead to ESRD in 30% to 50% of patients after 25 years or
renal failure. However, the possibility of recurrence of the more of follow-up. IgAN is one of the most common recurrent
original disease after transplantation was described in a semi- GN after transplantation (6).
nal paper more than 40 years ago (1), and it is now clear that all A review of the literature reported that recurrences develop
forms of GN may recur after kidney transplantation and po- in approximately 33% of patients with large differences, rang-
tentially jeopardize the long-term outlook for graft survival ing between 9% and 61%, among the different series (7). Several
(2,3), although the risk of recurrence is different for the various reasons can account for these large discrepancies. The indica-
categories of primary GN. Recent studies provided new clues tions for undergoing a graft biopsy were variable in the differ-
for better understanding the pathogenesis and thereby enhanc- ent studies and it is likely that several patients with asymptom-
ing the possibility for improving the prevention or treatment of atic hematuria and/or proteinuria never received biopsy. The
some forms of recurrent GN. This review will focus on the four follow-ups were quite different and the risk of recurrence was
more frequent forms of primary (idiopathic) GN that can recur probably underestimated for patients followed for short times.
after renal transplantation in Western countries; namely, IgA Longer follow-up times yield higher recurrence rates. There are
nephritis (IgAN), focal segmental glomerulosclerosis (FSGS), different racial and geographical distributions of IgAN. In some
membranous nephropathy (MN), and membranoproliferative cases, IgA deposits could be already present in the donated
GN (MPGN). Recurrence of secondary forms of glomerular kidneys. As an example, in a Japanese series, mesangial IgA
diseases will not be discussed, nor will the uncommon event of deposits were present in 16% of 510 donated kidneys (8), al-
recurrent anti-neutrophil cytoplasmic antibody-positive small- though it is not clear if the deposited IgA were abnormally
vessel vasculitis in renal allografts. glycosylated.
Recurrences of IgAN can be “immunopathologic” only; dis-
IgAN covered on a “protocol” renal biopsy in an asymptomatic pa-
IgAN is a primary GN characterized by the dominant or tient; or “clinical,” which is delineated by abnormalities in the
co-dominant diffuse mesangial deposition of IgA, usually IgA1. urine and/or renal function leading to a renal biopsy. Clinical
The deposits consist of an aberrant galactose-deficient form of recurrence of IgAN may occur immediately after transplanta-
tion, but on average the diagnosis is made approximately 3
years after transplantation (7,9) and is usually heralded by
Published online ahead of print. Publication date available at www.cjasn.org. hematuria and low-grade proteinuria. The results of renal
Correspondence: Dr. Richard J. Glassock, 8 Bethany, Laguna Niguel, CA 92677. transplantation in patients with IgAN have been differently
Phone: (949) 388-8885; Fax: (949) 388-8882; E-mail: Glassock@cox.net estimated. Some investigators (10,11) reported a more favorable

Copyright © 2010 by the American Society of Nephrology ISSN: 1555-9041/512–2363


2364 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 5: 2363–2372, 2010

outcome of the renal allograft in patients with IgAN than in anti-thymocyte globulins could be related to an augmented
other transplant recipients, but others found that graft survival production of regulatory T cells.
in an IgAN group was similar to that of non-IgAN renal trans- The finding that some apparently normal donors (living or
plant recipients (12–14). An analysis of the Eurotransplant reg- deceased) may have “hidden” IgA deposits in the kidney (9)
istry data reported the outcome of 1207 patients with IgAN and may have important relevance for pathogenesis of the recurrent
7935 other renal transplant recipients. The 10-year death-cen- disease. It may be presumed that the circulating autoantibody
sored graft survival was identical in the control group with and to exposed N-acetylgalactosamine residues on galactose-defi-
without the HLA-B8, DR3 haplotype (71.1% versus 70.2%) but cient IgA1 in some subjects with IgAN could react with the
significantly worse in IgAN patients carrying the HLA-B8, DR3 “planted” IgA deposits (posited also to be deficient in galactose
haplotype than in patients without it (52.5% versus 69.1%, P ⫽ residues thus exposing the neo-N-acetylgalactosamine epitopes)
0.009), suggesting that this HLA haplotype may represent a risk in the allograft and thus promote an immunopathological recur-
factor for transplant recipients with IgAN (15). The limit of this rence (4,6,30,31). Whether this immunopathological recurrence
study rests on the large heterogeneity between patients with would rise to the level of clinical detection would depend on
IgAN and the control population. A case-control study com- the activation of accessory factors (e.g., complement and cyto-
pared the long-term outcome of 106 patients transplanted be- kines [32]). Circulating immune complexes composed of IgG or
cause of IgAN with that of 212 non-IgAN patients transplanted IgA autoantibodies to galactose-deficient IgA1 and the relevant
in the same period and in the same center (16). After censoring autoantigen might theoretically participate in recurrent disease,
the graft failures in the first 6 months, the 10-year graft survival especially if these immune complexes escape normal reticulo-
probability was not significantly different between the two endothelial-dependent removal mechanisms (33).
groups: 75% in patients with IgAN and 82% in controls. The No specific therapy for recurrent IgAN is currently available.
regression coefficient of the reciprocal of plasma creatinine was A review of the U.S. Renal Data System examined the effect of
also similar in the two groups. In a Cox proportional hazards immunosuppressive medication on allograft failure due to re-
model analysis, IgAN recurrence was not associated with an current GN. After adjusting for important covariates, the use of
increased risk of graft failure. Other series reported that recur- cyclosporine, tacrolimus, azathioprine, mycophenolate mofetil,
rence of IgA deposition has only a modest and inconsistent sirolimus, or prednisone was not associated with graft failure
effect on graft survival. In the Australia-New Zealand registry, due to recurrent GN for any type of GN, including IgAN (34).
the long-term graft survival was not affected by the increased Angiotensin converting enzyme inhibitors (ACEIs) may be pre-
risk of recurrence in zero-mismatched kidneys (17). In two scribed because a study showed that their use could reduce
large Asian reports, the risk of graft loss due to recurrence proteinuria and blood pressure in transplant recipients with
ranged around 3% to 4% (18,19). However, in the very long IgAN (35). However, in using these drugs one should take into
term it is possible that recurrent IgAN may represent a sub- account that the use of ACEIs or angiotensin receptor blockers
stantial risk factor for graft failure. Rarely, a rapidly progressive (ARBs) in renal transplant recipients may be associated not
renal failure caused by an underlying crescentic GN may occur only with reduction in proteinuria but also with significant
(20,21). decreases in GFR and hematocrit (36). Tonsillectomy has been
Recurrence tends to occur more frequently in younger pa- suggested to be of benefit by Japanese investigators. Sixteen of
tients and in those with a rapid progression of the original 28 transplant patients with biopsy-proven recurrence of IgAN
disease (16,22,23). It is unclear if recurrence is (24,25) or is not and persistent proteinuria underwent tonsillectomy alone,
(16,26) more frequent in transplants using related donors. A whereas the remaining 12 patients did not receive tonsillec-
review of the data collected by the Australia-New Zealand tomy. Proteinuria decreased significantly in all of the tonsillec-
registry reported that biopsy-proven IgAN recurrence was sig- tomized patients but in none of the other group. The reduction
nificantly more frequent in zero HLA-mismatched living donor in proteinuria after tonsillectomy was especially marked in
grafts (17). However, these results should be interpreted with patients with mild mesangial changes on renal biopsy (37).
caution because several details are lacking in retrospective re- Long-term outcomes were not examined in this study. In the
views of registry data. Most other pretransplant characteristics rare cases of recurrent IgAN associated with a rapidly progres-
are not consistently predictive for recurrence, although some sive course and crescents at biopsy, a trial with high-dose
authors have suggested that latent IgA deposits in the donor corticosteroids, cyclophosphamide, and plasmapheresis may be
kidney (in “zero” time biopsies of the transplanted kidney) are attempted, although the results are usually poor.
highly associated with a recurrence of disease (27), and under-
lying crescentic GN with rapid progression to ESRD may also FSGS
predict a higher risk for recurrence (28). There is now general Idiopathic FSGS is often associated with a fully developed
agreement that the type and intensity of posttransplant immu- nephrotic syndrome (NS) and may affect children and adults.
nosuppression does not influence the risk of IgAN recurrence. In the absence of a treatment-induced remission, FSGS com-
However, Berthoux et al. (29) recently reported that the 10-year monly progresses to ESRD. Approximately 30% of patients
cumulative recurrence rate of IgAN was 9% (3 of 29) in patients develop recurrence of FSGS in the first kidney allograft (38).
who received induction with anti-thymocyte globulins versus The risk of recurrence with a second graft in patients who lost
41% (21 of 52) in patients who did not receive such induction a first graft because of recurrence may approach 100% (39). Two
therapy. The authors hypothesized that the protective effect of patterns of clinical presentations of recurrent FSGS after trans-
Clin J Am Soc Nephrol 5: 2363–2372, 2010 Recurrent Glomerulonephritis 2365

plantation are recognized: (1) an early recurrence (the most approximately 8% (52). Therefore, caution should be observed
frequent) characterized by a massive proteinuria within hours in transplanting patients with NPHS2 mutations using the kid-
to days after implantation of the new kidney, and (2) a late ney of their parents who are obligate carriers of the NPHS2
recurrence that develops insidiously several months or years mutation (53). It is also possible to speculate that in patients
after transplantation. with genetic FSGS the transplant of a normal kidney may evoke
The graft survival in patients with FSGS is lower for children an alloimmune response to slit diaphragm proteins.
than adults, particularly if the patient is white or Hispanic The frequent occurrence of a rapid relapse of proteinuria
(40,41), and is lower in children with FSGS than in those with after transplantation led to a postulate that a circulating factor,
other renal diseases (42,43). The renal prognosis is strongly perhaps secreted by an abnormal clone of T cells, may induce
influenced by recurrence because the relative risk of graft fail- redistribution and loss of nephrin and podocin, two critical
ure is 2.25 times higher in patients with recurrent FSGS as components of the glomerular slit-diaphragm of podocytes,
compared with patients without recurrence (2). Graft loss thereby reducing the efficiency of the glomerular barrier to
caused by recurrent FSGS is significantly higher in children proteins. However, despite intensive research by different
receiving living donor transplants compared with deceased groups of investigators (54 –58), the biochemical nature of the
donor transplants in children (44). Only few single-center stud- posited circulating permeability factor(s) remains unknown,
ies reported data on graft survival in adults with FSGS. Pardon and we still lack a reliable chemical or immunoassay for its
et al. (45) reported a graft survival rate of 73% at 5 years in 33 presence and quantitation.
transplanted adults with FSGS, with a significant difference The management of patients with recurrent FSGS and NS is
between the patients with (57%) or without (82%) recurrence. difficult, controversial, and none of the multiple approaches
No comparison with a control group was made. Moroni et al. currently available has been shown to be consistently benefi-
(46) compared the long-term outcomes of 52 renal transplants cial. Perhaps more targeted approaches will emerge based on
performed in adults with FSGS with those of 104 matched our improving knowledge of podocyte biology and function.
controls. At 15 years, graft survival was not significantly dif- An amelioration of proteinuria has been reported in children
ferent: 56% in FSGS and 64% in controls. Recurrence of FSGS treated with intravenous cyclosporine at high doses (59 – 62).
occurred in 12 patients (23%) and led to graft failure in 7 within This anti-proteinuric effect of cyclosporine may be attributed to
10 months (median). In the other five patients, proteinuria suppression of T cells and inhibited production of their cyto-
remitted and grafts were functioning 106 months (median) after kines and to the inhibition of calcineurin-mediated dephos-
transplantation. In the long term, patient and renal allograft phorylation of synaptopodin, a protein critical for stabilizing
survivals of adults with FSGS were comparable to those of the actin cytoskeleton in kidney podocytes (63). Cyclosporine
controls. and other calcineurin inhibitors thus have systemic immuno-
Several clinical factors have been reported to be associated regulatory and local podocyte modulatory actions: Either one
with an increased risk of recurrence, including young age, or both can be responsible for their anti-proteinuric effects in
mesangial proliferation in the native kidneys, a rapid progres- recurrent FSGS. However, the long-term efficacy and tolerance
sion to ESRD, a pretransplant bilateral nephrectomy, white of such a therapy remains to be established. The most com-
ethnicity, and specific aspects of genetic background (47). The monly used therapeutic approach is the use of plasma exchange
histologic variant type of FSGS observed in the native kidneys (PE) or immunoadsorption with protein A (59,64 – 66). A review
does not seem to reliably predict either recurrence or type of of the literature reported that 70% of children and 63% of adults
FSGS seen on the allograft (48). There is a higher risk of recur- with recurrent FSGS who received PE entered complete or
rence in living donor transplant pediatric recipients (49); how- partial remission of proteinuria (47). However, all studies were
ever, the reduced risk of rejection and a lower immunosuppres- retrospective, uncontrolled, and most of them had only short-
sion in living-related transplants may overcome the deleterious term follow-ups. In a pilot trial, 10 adults with FSGS recurrence
effect of recurrent GN. A review of the U.S. Renal Data System received concomitantly high-dose steroids, intravenous cyclo-
data reported that, after correction for other factors, living sporine for 14 days followed by oral cyclosporine therapy, and
donor transplants had no association with graft loss from re- an intensive and prolonged course of PE. Complete, rapid, and
current FSGS; rather, a living donor transplant was associated sustained remission was obtained in 9 of 10 patients. At month
with superior overall graft survival (50). Cibrik and co-workers 3 and month 12, proteinuria was 0.16 and 0.19 g/d, respec-
(51) also found that the risk of death-censored graft loss was 1% tively. PE was stopped at month 9 in all patients except for one
per year in adults with FSGS who received a zero HLA mis- patient who remained PE dependent (64). A protective role of
match kidney from living donors versus a 4.4% loss per year for prophylactic PE before transplantation has also been reported
patients with FSGS who received a zero HLA mismatch kidney (65,66). Several single-case reports pointed out the benefit of
from deceased donors. Another issue concerns the living-re- rituximab when given alone or in combination with PE (67–72),
lated kidney donation to a subject affected by familial FSGS. but failures were also reported (73–76). Currently, PE combined
Until recently the current opinion was that patients with famil- with high-dose calcineurin inhibitors with or without rituximab
ial FSGS caused by mutations of NPHS2, the gene encoding seems to be the most promising approach, but further con-
podocin, did not run any risk of recurrence after transplanta- trolled trials are needed to define the optimal therapeutic reg-
tion. However, there is now evidence that the risk of recurrence imens to treat recurrence of FSGS (77).
in patients with the NPHS2 mutation is low (but not zero) at Despite the risk of recurrence, patients with FSGS should not
2366 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 5: 2363–2372, 2010

be excluded from transplantation. In the case of living dona- nificant difference in death, graft failures, or serum creatinine
tion, the possibility of recurrence and its consequences should between IMN with recurrent disease and those without recur-
be clearly explained to the donor and the recipient and pre- rence after mean follow-ups of 73 and 57 months, respectively
emptive PE should be planned in advance of transplantation. (108).
An early and aggressive treatment should be provided if pro- The mechanisms leading to IMN recurrence are still far from
teinuria develops. being elucidated. There is now evidence that MN is triggered
by autoantibodies directed against podocyte proteins. Debiec et
IMN al. (109) identified in newborns from mothers deficient in these
IMN is characterized histologically by uniform thickening of podocyte proteins circulating specific antibodies (IgG4 and
the glomerular capillary due to immune-complex deposits in IgG1) against neutral endopeptidase. This is an example of an
the outer or subepithelial aspect of the glomerular basement allo-immune reaction causing MN and may be more akin to the
membrane. IMN is a frequent cause of NS in adults and may pathogenesis of de novo MN. Recently, circulating autoantibod-
lead in 40% to 50% of patients to ESRD in the long term (78,79). ies directed against other podocyte enzymes, such as M-type
A recurrence of IMN after renal transplantation is probably phospholipase-2 receptors (110) and aldose reductase and man-
more frequent than generally estimated. Up to now, almost 300 ganese superoxide dismutase (111), have been detected in
cases of recurrent MN have been reported in renal transplant adults with IMN and are uniformly absent in secondary forms
patients (80 –108), most cases occurring in adults. However, the of MN. The expression of these autoantigens in podocytes can
true proportion of recurrence is difficult to assess because the trigger the production of specific antibodies (mainly IgG4 and
indications for graft biopsy are extremely variable among trans- IgG1) and in situ deposits of immune complexes on the epithe-
plant centers. Moreover a de novo form of secondary MN may lial side of the glomerular basement membrane, with conse-
develop in transplanted kidneys showing a histologic pattern quent activation of complement, oxygen radicals, and other
indistinguishable from recurrent IMN. This de novo MN may components of the inflammatory response. As far as the patho-
occur even more frequently than the recurrent MN. In a study genesis of recurrent IMN is concerned, it is possible to hypoth-
with protocol biopsies, 6 of 17 cases of MN were identified; of esize that recurrent disease is due to the reaction of autoanti-
them 6 (35%) were considered to be recurrent IMN and 11 bodies (anti-phospholipase A2 receptor) from the circulation
(65%) de novo MN (108). With these difficulties in mind, the rate with conformational epitopes exposed or genetically deter-
of IMN recurrence has been estimated to range between 30% mined in the donor kidney. This possibility has been recently
and 44% (96,108). confirmed by clinical observations (112).
Recurrence of IMN is usually diagnosed between the 2nd Symptomatic treatment with diuretics, ACEIs, ARBs, hypo-
and 3rd year after transplantation, but earlier and later cases lipemic drugs, and anticoagulants may help in reducing the
have been described. So far, no clinical or histologic factor signs and symptoms related to the NS in recurrent IMN. No
seems to reliably predict the risk of recurrence. In one series convincing evidence exists that corticosteroids, cytotoxic drugs,
(89) posttransplant recurrence occurred earlier in recipients of a or other immunosuppressive agents are of benefit in recurrent
living-donor allograft (mean 9.3 months after transplantation) IMN. Rituximab has shown very promising effects in patients
than in deceased donor transplant recipients (18.2 months). with IMN in native kidneys (113,114) and has also been used
However, a study from Hong Kong reported that MN (includ- successfully in anecdotal cases of posttransplant IMN recur-
ing recurrent IMN and de novo MN) occurred later, at a mean of rence (100,103,104). In a recent study, eight patients with recur-
45 months after transplantation (98). The initial clinical mani- rent IMN and a mean proteinuria of 4.5 g/d were given two
festations of recurrent IMN may be mild or absent, and in separate doses of intravenous rituximab (1000 mg each) 2
several patients recurrence could be detected only by protocol weeks apart. Twelve months later, 35% of patients had a com-
renal biopsies (101). However, many patients show a progres- plete remission and another 40% had a partial remission. After
sive increase in proteinuria over time and can eventually de- 24 months, one patient had relapsed. Post-treatment biopsies
velop a full-blown NS. Proteinuria may spontaneously improve showed reabsorption of electron-dense immune deposits in six
or even disappear only rarely (94,102). of seven patients (107). In another series, four patients with
The results of renal transplantation in patients with IMN are recurrent IMN and severe proteinuria were given rituximab,
not different than those observed in patients with other renal either at a weekly dose of 375 mg/m2 for 4 weeks, or at a dose
diseases, including primary GN. The Australian registry re- of 100 mg repeated 2 weeks later. Two patients responded to
ported that the actuarial renal allograft survival at 10 years was the first course and the other two patients received a second
similar in patients transplanted because of IMN and in 1505 course of rituximab. Mean proteinuria decreased from 4.0 to
transplanted patients with other renal diseases (3). The out- 1.8 g per day, and serum creatinine remained stable at approx-
come of recurrent IMN may be as variable as in the original imately 1.5 mg/dl (108).
native disease and its effect on graft survival has been differ-
ently estimated. Some studies have reported that 60% to 65% of MPGN Types I and II (Dense Deposit
patients with recurrent IMN progressed to ESRD in a mean of Disease)
4 years after diagnosis (89,95). However, in many patients the MPGN is “pattern of injury” rather than a disease (115). It is
graft failure was caused by rejection rather than recurrence of now known to have a very diverse array of underlying causes,
IMN. Other investigators reported that there was not any sig- and the group designated as ‘idiopathic” MPGN has corre-
Clin J Am Soc Nephrol 5: 2363–2372, 2010 Recurrent Glomerulonephritis 2367

spondingly declined in size (115). Nevertheless. MPGN is com- MPGN and contribute to recurrent disease (115,116), frequently
mon cause of recurrent GN in allografts. The reported rate of appearing within a few months after transplantation (116).
recurrence of MPGN has been quite variable (27% to 65%) (116). Finally, a newly described entity, known as “GN C3,” may also
MPGN has previously been divided into subtypes (types I, II, have clinical features similar to DDD and type I MPGN, but it
III, etc.) on the basis of ultrastructural features. Type I MPGN lacks the ultrastructural features of typical DDD and fails to
characteristically has subendothelial electron-dense deposits show any immune complex deposits characteristically found in
containing IgG and C3, which presumably represent immune type I MPGN. Only C3 without IgG deposits are observed.
complexes. Type II MPGN (also known as dense deposit dis- Deficiencies of complement factor H, I, or membrane cofactor
ease [DDD]) shows an electron-dense transformation of the protein are frequently noted. The risk of recurrence of GN C3 in
glomerular basement membrane and deposition of C3 (and the allograft is not well known but would be presumed to be
other complement components without IgG deposition). In elevated if abnormalities in complement dysregulation are
previous series, types I and II MPGN (DDD) were considered found.
together, whereas the current trend is to separate DDD as a The successful treatment of an established recurrence of
unique clinicopathologic entity having a higher risk of recur- DDD is problematical, so prevention and anticipatory manage-
rence than typical type I MPGN (117). Differences in recurrence ment based on precise assessment of the underlying mecha-
rates between type I MPGN and DDD may relate more to the nism responsible for the MPGN is very important. Patients with
superimposition of crescents than to the underlying ultrastruc- complement dysregulation (e.g., factor H deficiency) should
tural features (118). Risk of recurrence may be marginally receive replacement infusions (fresh frozen plasma) before and
higher in living related donors. The most important aspect of after grafting (123), although no controlled trials of the efficacy
recurrent MPGN or DDD is the identification of the processes of this approach have yet been conducted. Plasma therapy has
responsible for the original native disease. Recurrent MPGN shown very promising results in atypical hemolytic uremic
due to lupus nephritis, monoclonal gammopathies, thrombotic syndrome associated with genetic mutations in factor H
microangiopathies, and more frequently HCV infection (119) (124,125). PE (with fresh frozen plasma replacement) and/or
do occur, but they are beyond the scope of this review. rituximab might also be helpful in patients with a neutralizing
Recurrent MPGN type I can have significant deleterious ef- autoantibody to factor H. Eculizumab (a monoclonal antibody
fects on graft survival, especially when superimposed extensive to C5a) may also be beneficial, but this has not been yet been
crescentic disease is present (118), and thus should be pre- shown in a randomized trial (123). Patients with genetic causes
vented in so far as is possible by careful pretransplant evalua- for factor H or I deficiency may require combined liver and
tion. kidney transplantation to avoid recurrences (125), but this is
Intensification of immunosuppressive therapy in recurrent mainly derived from experience with hereditary forms of atyp-
MPGN can be hazardous because it may lead to overimmuno- ical hemolytic uremic syndrome also associated with factor H
suppression and has little documented effect on the outcome of deficiency (126). Patients with documented underlying mono-
the recurrence, except perhaps when extensive crescentic dis- clonal gammopathies should receive appropriate aggressive
ease is present. Occasional anecdotes and small series have chemotherapy and/or autologous stem cell transplantation be-
suggested that improvement may be seen in recurrent MPGN fore kidney grafting so as to minimize the risk of recurrence.
type I with cyclophosphamide (120) or high-dose mycopheno-
late mofetil (121), but no controlled trial has yet confirmed the
efficacy or safety of these approaches. Treatment of truly “id-
Conclusions
Although recurrent GN is relatively frequent and may
iopathic” recurrent MPGN is generally very disappointing and
worsen the outcome of renal allograft in some patients, its effect
graft loss due to recurrence is common (116).
on the fate of a renal allograft is diluted by several other risk
factors, including death with functioning graft, the quality of
DDD the donated kidney, rejection, drug nephrotoxicity, infections,
This disease has a very high risk of recurrence (approaching
and other renal and extrarenal complications such as hyperten-
100%). Most patients have low serum C3 levels, and 70% to 80%
sion, diabetes, coronary artery disease, liver disease, and can-
also have a circulating autoantibody to C3Bb known as C3
cer. These factors may have a greater effect than recurrent GN
nephritic factor (C3Nef). Some patients with DDD may also
on long-term graft survival, as demonstrated by large series
have an abnormality in complement cascade regulation such as
showing that overall long-term graft survival is similar in re-
deficiencies of factor H) (122). Thus, hypocomplementemia
cipients with many forms of primary GN and in those with
and/or isolated C3 deposits suggesting DDD in a native kidney
other renal diseases (2,127).
biopsy showing a pattern of MPGN is a feature highly associ-
ated with risk for recurrence (115,116), When ultrastructural
analysis of the native kidney biopsy reveals DDD an evaluation Disclosures
for complement dysregulation (factor H, I, or membrane cofac- None.
tor protein levels) seems appropriate. Serum C3 nephritic factor
levels do not reliably predict the risk of recurrence of DDD in
the renal allograft. Subtle monoclonal Ig deposition diseases References
such as light-chain deposition disease can also mimic idiopathic 1. Glassock RJ, Feldman D, Reynolds ES, Dammin GJ, Merrill
2368 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 5: 2363–2372, 2010

JP: Human renal isografts: A clinical and pathologic anal- 19. Ng YS, Vathsala A, Chew ST, Chiang GS, Woo KT: Long-
ysis. Medicine (Baltimore) 47: 411– 454, 1968 term outcome of renal allografts in patients with immuno-
2. Hariharan S, Adams MB, Brennan DC, Davis CL, First RM, globulin A nephropathy. Med J Malaysia 62: 109 –113, 2007
Johnson CP, Ouseph R, Peddi VR, Pelz CJ, Roza AM, 20. Benabdallah L, Rerolle JP, Peraldi MN, Noël LH, Bruneel
Vincenti F: Recurrent and de novo glomerular disease after MF, Carron PL, Morelon E, Kreis H: An unusual recur-
renal transplantation. A report from the Renal Allograft rence of crescentic nephritis after renal transplantation for
Disease Registry (RADR). Transplantation 68: 635– 641, 1999 IgA nephropathy. Am J Kidney Dis 40: E20, 2002
3. Briganti EM, Russ GR, McNeil JJ, Atkins RC, Chadbans J: 21. Kowalewska J, Yuan S, Sustento-Reodica N, Nicosia RF,
Risk of renal allograft loss from recurrent glomerulone- Smith KD, Davis CL, Alpers CE: IgA nephropathy with
phritis. N Engl J Med 347: 103–109, 2002 crescents in kidney transplant recipients. Am J Kidney Dis
4. Novak J, Julian B, Tomana M, Mestecky J: IgA glycosyla- 45: 167–175, 2005
tion and IgA immune complexes in the pathogenesis of 22. Chailimpamontree W, Dmitrienko S, Li G, Balshaw R,
IgA nephropathy. Semin Nephrol 28: 78 – 87, 2008 Magil A, Shapiro RJ, Landsberg D, Gill J, Keown PA;
5. Barratt J, Eitner F, Feehally J, Floege J: Immune complex Genome Canada Biomarkers in Transplantation Groupe:
formation in IgA nephropathy: A case of the “right” anti- Probability, predictors, and prognosis of posttransplanta-
bodies in the “wrong” place at the “wrong” time? Nephrol tion glomerulonephritis. J Am Soc Nephrol 20: 843– 851, 2009
Dial Transplant 24: 3620 –3623, 2009 23. Floege J: Recurrent IgA nephropathy after renal transplan-
6. Furness PN, Roberts ISD, Briggs JD: Recurrent glomerular tation. Semin Nephrol 24: 287–291, 2004
disease in transplants. Transplant Proc 34: 242, 2002 24. Andresdottir MB, Haasnot GW, Doxiadis II, Persjin GG,
7. Ponticelli C, Traversi L, Banfi G: Renal transplantation in Claas FH: Exclusive characteristics of graft survival and
patients with IgA mesangial glomerulonephritis. Pediatr risk factors in recipients of immunoglobulin A nephropa-
Transplant 8: 334 –338, 2004 thy: A retrospective analysis of registry data. Transplanta-
8. Suzuki K, Honda K, Tanabe K, Toma H, Nihei H, Yamagu- tion 80: 1012–1018, 2005
chi Y: Incidence of latent mesangial IgA deposition in renal 25. Wang AYM, Lai FM, Yu AWY Lam PK, Chow KM, Choi
allograft donors in Japan. Kidney Int 63: 2286 –2294, 2003 PC, Lui SF, Li PK: Recurrent IgA nephropathy in renal
9. Hariharan S, Savin VJ: Recurrent and de novo disease after transplant allografts. Am J Kidney Dis 38: 588 –596, 2001
renal transplantation: A report from the Renal Allograft 26. Kim YS, Moon JI, Jeong HJ, Kim MS, Kim SI, Choi KH, Lee
Disease Registry. Pediatr Transplant 8: 349 –350, 2004 HY, Han DS, Park K: Live donor renal allograft in end-
10. Lim EC, Chia D, Terasaki PI: Studies of sera from IgA stage renal failure patients from immunoglobulin A ne-
nephropathy patients to explain high kidney graft sur- phropathy. Transplantation 71: 233–238, 2001
vival. Hum Immunol 32: 81– 86, 1991 27. Moriyama T, Nitta K, Suzuki K, Honda K, Horita S, Uchida
11. Jeong HJ, Park SK, Cho YM, Kim MS, Kim YS, Choi J, Kim K, Yumura W, Tanabe K, Toma H, Nihei H, Yamaguchi Y:
SI, Lim BJ: Progression of renal allograft histology after Latent IgA deposition from donor kidney is the major risk
renal transplantation in recurrent and nonrecurrent immu- factor for recurrent IgA nephropathy in renal transplanta-
noglobulin A nephropathy. Hum Pathol 39: 1511–1518, 2008 tion. Clin Transplant 19[Suppl 14]: 41– 48, 2005
12. Bumgardner GL, Amend WC, Ascher NL, Vincenti FG: 28. Tang Z, Ji SM, Chen DR Wen JQ, Liu ZH, Li LS: Recurrent
Single-center long-term results of renal transplantation for or de novo IgA nephropathy with crescent formation after
IgA nephropathy. Transplantation 65: 1053–1060, 1998 renal transplantation. Ren Fail 30: 611– 616, 2008
13. Soler MJ, Mir M, Rodriguez E, Orfila A, Munne A, Vázquez 29. Berthoux F, El Deeb S, Mariat C, Diconne E, Laurent B,
S, Lloveras J, Puig JM: Recurrence of IgA nephropathy and Thibaudin L: Anti-thymocyte globulin (ATG) induction
Henoch–Schoenlein purpura after kidney transplantation: therapy and disease recurrence in renal transplant recipi-
Risk factors and graft survival. Transplant Proc 37: 3705– ents with primary IgA nephropathy. Transplantation 85:
3709, 2005 1505–1507, 2008
14. Chacko B, George JT, Neelakantan N, Korula A, Chakko 30. Suzuki H, Fan R, Zhang Z, Brown R, Hall S, Julian BA,
JK: Outcomes of renal transplantation in patients with Chatham WW, Suzuki Y, Wyatt RJ, Moldoveanu Z, Lee JY,
immunoglobulin A nephropathy in India. J Postgrad Med Robinson J, Tomana M, Tomino Y, Mestecky J, Novak J:
53: 92–95, 2007 Aberrantly glycosylated IgA1 in IgA nephropathy patients
15. Andresdottir MB, Haasnoot GW, Persijn GG, Claas FH: is recognized by IgG antibodies with restricted heteroge-
HLA-B8, DR3: A new risk factor for graft failure after renal neity. J Clin Invest 119: 1666 –1677, 2009
transplantation in patients with underlying immunoglob- 31. Glassock RJ: Analyzing antibody activity in IgA nephrop-
ulin A nephropathy. Clin Transplant 23: 660 – 665, 2009 athy. J Clin Invest 119: 1450 –1452, 2009
16. Ponticelli C, Traversi L, Feliciani A, Cesana BM, Banfi G, 32. Coppo R, Feehally J, Glassock RJ: IgA nephropathy at two
Tarantino A: Kidney transplantation in patients with IgA score and one. Kidney Int 77: 181–186, 2010
mesangial glomerulonephritis. Kidney Int 60: 1948 –1954, 33. Mestecky J, Suzuki H, Yanagihara T, Moldoveneanu Z,
2001 Tomana M, Matousovic C, Julian BA, Novak J: IgA ne-
17. McDonald SP, Russ GR: Recurrence of IgA nephropathy phropathy: Current views of immune complex formation.
among renal allograft recipients from living donors is Contrib Nephrol 157: 58 – 63, 2007
greater among those with zero HLA mismatches. Trans- 34. Mulay AV, van Walraven C, Knoll GA: Impact of immu-
plantation 27, 759 –762, 2006 nosuppressive medication on the risk of renal allograft
18. Choy BY, Chan TM, Lo SK, Lo WK, Lai KN: Renal trans- failure due to recurrent glomerulonephritis. Am J Trans-
plantation in patients with primary immunoglobulin A plant 9: 804 – 811, 2009
nephropathy. Nephrol Dial Transplant 18: 2399 –2404, 2003 35. Oka K, Imai E, Moriyama T, Akagi Y, Ando A, Hori M,
Clin J Am Soc Nephrol 5: 2363–2372, 2010 Recurrent Glomerulonephritis 2369

Okuyama A, Toki K, Kyo M, Kokado Y, Takahara S: A rent focal segmental glomerulosclerosis in renal transplant
clinicopathological study of IgA nephropathy in renal recipients in the United States. Am J Kidney Dis 237: 366 –
transplant recipients: Beneficial effect of angiotensin-con- 373, 2001
verting enzyme inhibitor. Nephrol Dial Transplant 15: 689 – 51. Cibrik DM, Kaplan B, Campbell DA, Meier-Kriesche HU:
695, 2000 Renal allograft survival in transplant recipients with focal
36. Hiremath S, Fergusson D, Doucette S, Mulay AV, Knoll segmental glomerulosclerosis. Am J Transplant 3: 64 – 67,
GA: Renin angiotensin system blockade in kidney trans- 2003
plantation: A systematic review of the evidence. Am J 52. Weber S, Gribouval O, Esquivel EL, Morinière V, Tête MJ,
Transplant 7: 2350 –2360, 2007 Legendre C, Niaudet P, Antignac C: NPHS2 mutation anal-
37. Kennoki T, Ishida H, Yamaguchi Y, Tanabe K: Proteinuria- ysis shows genetic heterogeneity of steroid-resistant ne-
reducing effects of tonsillectomy alone in IgA nephropathy phrotic syndrome and low post-transplant recurrence. Kid-
recurring after kidney transplantation. Transplantation 88: ney Int 66: 571–579, 2004
935–941, 2009 53. Vincenti F, Ghiggeri GM: New insights into the pathogen-
38. Couser W: Recurrent glomerulonephritis in the renal allo- esis and the therapy of recurrent focal glomerulosclerosis.
graft: An update of selected areas. Exp Clin Transplant 3: Am J Transplant 5: 1179 –1185, 2005
283–288, 2005 54. Savin VJ, Sharma R, Sharma M, McCarthy ET, Swan SK,
39. Newstead CG: Recurrent disease in renal transplants. Ellis E, Lovell H, Warady B, Gunwar S, Chonko AM,
Nephrol Dial Transplant 18[Suppl 6]: 68 –74, 2003 Artero M, Vincenti F: Circulating factor associated with
40. Tejani A, Stablein DH: Recurrence of focal segmental glo- increased glomerular permeability to albumin in recurrent
merulosclerosis posttransplantation: A special report of the focal segmental glomerulosclerosis. N Engl J Med 334: 878 –
North American Pediatric Renal Transplant Cooperative 883, 1996
Study. J Am Soc Nephrol 2[Suppl]: S258 –S263, 1992 55. Dantal J, Godfrin Y, Koll R, Perretto S, Naulet J, Bouhours
41. Fine RN: Recurrence of nephrotic syndrome/focal segmen- JF, Soulillou JP: Antihuman immunoglobulin affinity im-
tal glomerulosclerosis following renal transplantation in munoadsorption strongly decreases proteinuria in patients
children. Pediatr Nephrol 22: 496 –502, 2007 with relapsing nephrotic syndrome. J Am Soc Nephrol 9:
42. Carraro M, Caridi G, Bruschi M, Artero M, Bertelli R,
1709 –1715, 1998
Zennaro C, Musante L, Candiano G, Perfumo F, Ghiggeri
56. Rea R, Smith C, Sandhu K, Kwan J, Tomson C: Successful
GM: Serum glomerular permeability activity in patients
transplant of a kidney with focal segmental glomeruloscle-
with podocin mutation (NPHS2) and steroid resistant ne-
rosis. Nephrol Dial Transplant 16: 416 – 417, 2001
phrotic syndrome. J Am Soc Nephrol 13: 1946 –1952, 2002
57. Savin VJ, McCarthy ET, Sharma M: Permeability factors in
43. Fuentes GM, Meseguer CG, Carrion AP, Hijosa MM, Gar-
focal segmental glomerulosclerosis. Semin Nephrol 23: 147–
cia-Pose A, Melgar AA, Torres MN: Long-term outcome of
160, 2003
focal segmental glomerulosclerosis after pediatric renal
58. Trachtman H, Greenbaum LA, McCarthy ET, Sharma M,
transplantation. Pediatr Nephrol 25: 529 –534, 2010
Gauthier BG, Frank R, Warady B, Savin VJ: Glomerular
44. Baum MA: Outcomes after renal transplantation for FSGS
permeability activity: Prevalence and prognostic value in
in children. Pediatr Transplant 8: 329 –333, 2004
pediatric patients with idiopathic nephrotic syndrome.
45. Pardon A, Audard V, Caillard S, Moulin B, Desvaux D,
Bentaarit B, Remy P, Sahali D, Roudot-Thoraval F, Lang P, Am J Kidney Dis 44: 604 – 610, 2004
Grimbert P: Risk factors and outcome of focal and segmen- 59. Canaud G, Martinez F, Noel LH, Mamzer MF, Niaudet P,
tal glomerulosclerosis recurrence in adult renal transplant Legendre C: Therapeutic approach to focal and segmental
recipients. Nephrol Dial Transplant 21: 1053–1059, 2006 glomerulosclerosis recurrence in kidney transplant recipi-
46. Moroni G, Gallelli B, Quaglini S, Banfi G, Montagnino G, ents. Transplant Rev (Orlando) 24: 121–128, 2010
Messa P: Long-term outcome of renal transplantation in 60. Ingulli E, Tejani A: Incidence, treatment and outcome of
adults with focal segmental glomerulosclerosis. Transpl Int recurrent focal segmental glomerulosclerosis posttrans-
23: 208 –216, 2010 plantation in 42 allografts in children: A single center ex-
47. Ponticelli C: Recurrence of focal segmental glomeruloscle- perience. Transplantation 51: 401– 405, 1991
rosis (FSGS) after renal transplantation. Nephrol Dial Trans- 61. Salomon R, Gagnadoux MF, Niaudet P: Intravenous cyclo-
plant 25: 25–31, 2010 sporine therapy in recurrent nephrotic syndrome after
48. Canaud G, Dion D, Zuber J, Gubler MC, Sberro R, Thervet transplantation in children. Transplantation 75: 810 – 814,
E, Snanoudj R, Charbit M, Salomon R, Martinez F, Leg- 2003
endre C, Noel LH, Niaudet P: Recurrence of nephrotic 62. Raafat RH, Kalia A, Travis LB, Diven SC: High-dose cyclo-
syndrome after transplantation in a mixed population of sporine therapy for recurrent focal segmental glomerulo-
children and adults: Course of glomerular lesions and sclerosis in children. Am J Kidney Dis 44: 50 –56, 2004
value of the Columbia classification of histological variants 63. Faul C, Donnelly M, Merscher-Gomez S, Chang YH, Franz
of focal and segmental glomerulosclerosis (FSGS). Nephrol S, Delfgaauw J, Chang JM, Choi HY, Campbell KN, Kim K,
Dial Transplant 25: 1321–1328, 2010 Reiser J, Mundel P: The actin cytoskeleton of kidney podo-
49. Baum MA, Stablein DM, Panzarino VM Tejani A, Harmon cytes is a direct target of the antiproteinuric effect of cy-
WE, Alexander SR: Loss of living donor renal allograft closporine A. Nat Med 14: 931–938, 2008
advantage in children with focal segmental glomeruloscle- 64. Canaud G, Zuber J, Sberro R, Royale V, Anglicheau D,
rosis. Kidney Int 59: 328 –333, 2001 Snanoudj R, Gaha K, Thervet E, Lefrère F, Cavazzana-
50. Abbott KC, Sawyers ES, Oliver JD III, Ko CW, Kirk AD, Calvo M, Noël LH, Méjean A, Legendre C, Martinez F:
Welch PG, Peters TG, Agodoa LY: Graft loss due to recur- Intensive and prolonged treatment of focal and segmental
2370 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 5: 2363–2372, 2010

glomerulosclerosis recurrence in adult kidney transplant 79. Ponticelli C, Passerini P: Can prognostic factors assist ther-
recipients: A pilot study. Am J Transplant 9: 1081–1086, 2009 apeutic decisions in idiopathic membranous nephropathy?
65. Ohta T, Kawaguchi H, Hattori M, Komatsu Y, Akioka Y, J Nephrol 23: 156 –163, 2010
Nagata M, Shiraga H, Ito K, Takahashi K, Ishikawa N, 80. Crosson JT, Wathen RL, Raij L, Andersen RC, Andersen
Tanabe K, Yamaguchi Y, Ota K: Effect of pre-and postop- WR: Recurrence of membranous nephropathy after renal
erative plasmapheresis on posttransplant recurrence of fo- transplantation. Arch Int Med 135: 1101–1106, 1975
cal segmental glomerulosclerosis in children. Transplanta- 81. Petersen VP Olsen TS, Kissmeyer-Nielsen F, Bohman SO,
tion 71:628 – 633, 2001 Hansen ES, Skov PE, Solling K: Late failure of human renal
66. Gohh RY, Yango AF, Morrissey PE, Monaco AP, Gautam transplant. An analysis of transplant disease and graft
A, Sharma M, McCarthy ET, Savin VJ: Preemptive plasma- failure among 125 recipients surviving for one to eight
pheresis and recurrence of FSGS in high-risk renal trans- years. Medicine 54: 45– 69, 1975
plant recipients. Am J Transplant 5: 2907–2912, 2005 82. Morzycka M, Croker BP, Seigler HF, Tisher CC: Evaluation
67. Apeland T, Hartmann A: Rituximab therapy in early re- of recurrent glomerulonephritis in kidney allografts. Am J
current focal segmental sclerosis after renal transplanta- Med 72: 588 –598, 1982
tion. Nephrol Dial Transplant 23: 2091–2094, 2008 83. Berger BE, Vincenti F, Biava C, Amend WJ Jr, Feduska N,
68. Meyer TN, Thaiss F, Stahl RA: Immunoadsorbtion and Salvatierra O Jr: De novo and recurrent membranous glo-
rituximab therapy in a second living-related kidney trans- merulopathy following kidney transplantation. Transplan-
plant patient with recurrent focal segmental glomerulo- tation 35: 315–319, 1983
sclerosis. Transpl Int 20:1066 –1071, 2008 84. Obermiller LE, Hoy WE, Eversole M, Sterling WA: Recur-
69. Westphal S, Hansson S, Mjörnstedt L, Mölne J, Swerkers- rent membranous glomerulonephritis in two renal trans-
son S, Friman S: Early recurrence of nephrotic syndrome plants. Transplantation 40: 100 –102, 1985
(immunoglobulin m nephropathy) after renal transplanta- 85. Bansal VK, Kozeny GA, Fresco R, Vertuno LL, Hano JE: De
tion successfully treated with combinations of plasma ex- novo membranous nephropathy following renal transplan-
changes, immunoglobulin, and rituximab. Transplant Proc tation between conjoint twins. Transplantation 41: 404 – 406,
38: 2659 –2660, 2006 1986
70. Hristea D, Hadaya K, Marangon N, Buhler L, Villard J, 86. Montagnino G, Colturi C, Banfi G, Aroldi A, Tarantino A,
Morel P, Martin PY: Successful treatment of recurrent focal Ponticelli C: Membranous nephropathy in cyclosporine-
segmental glomerulosclerosis after kidney transplantation treated renal transplant recipients.Transplantation 47: 725–
by plasmapheresis and rituximab. Transpl Int 20: 102–105, 727, 1989.
2007 87. Truong L, Gelfand J, D’Agati V, Tomaszewski J, Appel G,
71. Dello Strologo L, Guzzo I, Laurenzi C, Vivarelli M, Parodi Hardy M, Pirani CL: De novo membranous nephropathy in
A, Barbano G, Camilla R, Scozzola F, Amore A, Ginevri F, renal allograft. A report of ten cases and review of the
Murer L: Use of Rituximab in focal glomerulosclerosis literature. Am J Kidney Dis 14: 131–141, 1989
relapses after renal transplantation. Transplantation 88: 88. Monga G, Mazzucco G, Basolo B, Quaranta S, Motta M,
417– 420, 2009 Segoloni G, Amoroso A: Membranous glomerulonephri-
72. Sakai K, Takasu J, Nihei H, Yonekura T, Aoki Y, tis (MGN) in transplanted kidneys: Morphologic inves-
Kawamura T, Mizuiri S, Aikawa A: Protocol biopsies for tigation on 256 renal allografts. Mod Pathol 6: 249 –258,
focal segmental glomerulosclerosis treated with plasma 1993
exchange and rituximab in a renal transplant patient. Clin 89. Josephson MA, Spargo B, Hollandsworth DO, Thistlewait
Transplant 24[Suppl 22]: 60 – 65, 2010 MD: The recurrence of membranous glomerulopathy in a
73. Kamar N, Faguer S, Esposito L, Guitard J, Nogier MB, renal transplant recipient: Case report and literature re-
Durand D, Rostaing L: Treatment of focal segmental glo- view. Am J Kidney Dis 24: 873– 878, 1994
merular sclerosis with rituximab: 2 case reports. Clin Neph- 90. Innes A, Woodrow G, Boyd SM, Beckingham IJ, Morgan
rol 67: 250 –254, 2007 AG: Recurrent membranous nephropathy in successive
74. El-Firjani A, Hoar S, Karpinski J, Bell R, Deschenes MJ, renal transplants. Nephrol Dial Transplant 9: 323–325,
Knoll GA: Post-transplant focal segmental glomeruloscle- 1994
rosis refractory to plasmapheresis and rituximab therapy. 91. Heidet L, Gagnadoux ME, Beziau A, Niaudet P, Broyer
Nephrol Dial Transplant 23: 425, 2008 M, Habib R: Recurrence of de novo membranous glomer-
75. Yabu JM, Ho B, Scandling JD, Vincenti F: Rituximab failed ulonephritis on renal grafts. Clin Nephrol 41: 314 –318,
to improve nephrotic syndrome in renal transplant pa- 1994
tients with recurrent focal segmental glomerulosclerosis. 92. Couchoud C, Pouteil-Noble C, Colon S, Touraine JL:
Am J Transplant. 8: 222–227, 2008 Recurrence of membranous nephropathy after renal
76. Rodríguez-Ferrero M, Ampuero J, Anaya F: Rituximab and transplantation. Transplantation 59: 1275–1279, 1995
chronic plasmapheresis therapy of nephrotic syndrome in 93. Odorico JS, Knechtle SJ, Rayhill SC, Pirsch JD,
renal transplantation patients with recurrent focal segmen- D’Alessandro AM, Belzer FO, Sollinger HW: The influ-
tal glomerulosclerosis. Transplant Proc 41: 2406 –2408, 2009 ence of native nephrectomy on the incidence of recurrent
77. Vinai M, Waber P, Seikaly MG: Recurrence of focal seg- disease following renal transplantation for primary glo-
mental glomerulosclerosis in renal allograft: An in-depth merulonephritis. Transplantation 61: 228 –234, 1996
review. Pediatr Transplant 14: 314 –325, 2010 94. Marcen R, Mampaso F, Teruel JL, Rivera ME, Orofino L,
78. Hogan SL, Muller KE, Jennette JC, Falk RJ: A review of Navarro-Antolin J, Ortuño J: Membranous nephropathy:
therapeutic studies of idiopathic membranous glomeru- Recurrence after kidney transplantation. Nephrol Dial
lopathy. Am J Kidney Dis 25: 862– 875, 1995 Transplant 11: 1129 –1133, 1996
Clin J Am Soc Nephrol 5: 2363–2372, 2010 Recurrent Glomerulonephritis 2371

95. Cosyns JP, Couchoud C, Pouteil-Noble C, Squifflet JP, endopeptidase antibodies. N Engl J Med 346: 2053–2060,
Pirson Y: Recurrence of membranous nephropathy after 2002
renal transplantation: Probability, outcome and risk fac- 110. Beck LH Jr, Bonegio RG, Lambeau G, Beck DM, Powell
tors. Clin Nephrol 50: 144 –153, 1998 DW, Cummins TD, Klein JB, Salant DJ: M-type phospho-
96. Lazowski P, Sablay LB, Glicklich D: Recurrent crescentic lipase A2 receptor as target antigen in idiopathic mem-
membranous nephropathy in two successive renal trans- branous nephropathy. N Engl J Med 361: 11–21, 2009
plants: Association with choroidal effusions and retinal 111. Prunotto M, Carnevali ML, Candiano G, Murtas C, Bruschi
detachment. Am J Nephrol 18: 146 –150, 1998 M, Corradini E, Trivelli A, Magnasco A, Petretto A, San-
97. Poduval RD, Josephson MA, Javaid B: Treatment of de novo tucci L, Mattei S, Gatti R, Scolari F, Kador P, Allegri L,
and recurrent membranous nephropathy in renal trans- Ghiggeri GM: Autoimmunity in Membranous Nephropa-
plant patients. Semin Nephrol 23: 392–399, 2003 thy Targets Aldose Reductase and SOD2. J Am Soc Nephrol
98. Chan KW, Chan GS, Tang S: Glomerular pathology of 21: 507–519, 2010
allograft kidneys in Hong Kong. Transplant Proc 37: 4293– 112. Stahl R, Hoxha E, Fechner K: PLA2R autoantibodies and
4296, 2005 recurrent membranous nephropathy after transplantation.
99. Carneiro-Roza F, Medina-Pestana JO, Moscoso-Solorzano N Engl J Med 363: 496 – 498, 2010
G, Franco M, Ozaki K, Mastroianni-Kirsztajn G: Initial 113. Remuzzi G, Chiurchiu C, Abbate M, Brusegan V, Bon-
response to immunosuppressive and renoprotective treat- tempelli M, Ruggenenti P: Rituximab for idiopathic
ment in posttransplant glomerulonephritis. Transplant Proc membranous nephropathy. Lancet 360: 923–924, 2002
38: 3491–3497, 2006 114. Fervenza FC, Cosio FG, Erickson SB, Specks U, Herzenberg
100. Gallon L, Chhabra D: Anti-CD20 monoclonal antibody AM, Dillon JJ, Leung N, Cohen IM, Wochos DN, Bergstralh
(rituximab) for the treatment of recurrent idiopathic mem- E, Hladunewich M, Cattran DC: Rituximab treatment of
branous nephropathy in a renal transplant patient. Am J idiopathic membranous nephropathy. Kidney Int 73: 117–
Transplant 6: 3017–3021, 2006 125, 2008
101. Dabade TS, Grande JP, Norby SM, Fervenza FC, Cosio FG: 115. Glassock RJ: Membranoproliferative glomerulonephritis.
Recurrent idiopathic membranous nephropathy after kid- In: Evidence-Based Nephrology, edited by Molony DA, Craid
ney transplantation: A surveillance biopsy study. Am J JC. London, Wiley-Blackwell, 2009:183–196
Transplant 8: 1318 –1322, 2008 116. Lorenz EC, Sethi S, Leung N, Dispenzieri A, Fervenza F,
102. Liebl R, Krämer BK: Recurrence and spontaneous remis- Cosio FG: Recurrent membranoproliferative glomerulone-
sion of membranous nephropathy after renal transplanta- phritis after kidney transplantation. Kidney Int 77: 721–728,
tion. Clin Nephrol 70: 419 – 421, 2008 2010
103. Weclawiak H, Ribes D, Guilbeau-Frugier C, Touchard G, 117. Braun MC, Stablein DM, Hamiwka LA, Bell L, Bartosh SM,
Kamar N, Mehrenberger M, Modesto A, Rostaing L: Re- Strife CF: Recurrence of membranoproliferative glomeru-
lapse of membranous glomerulopathy after kidney trans- lonephritis type II in renal allografts: the North American
plantation: Sustained remittance induced by rituximab. Pediatric Renal Transplant Cooperative Study Experience.
Clin Nephrol 69: 373–376, 2008 J Am Soc Nephrol 16: 2225–2233, 2005
104. Sirimongkolrat T, Premasathian N, Vongwiwatana A, Lim- 118. Little MA, Dupont P, Campbell E, Dorman A, Walshe JJ:
srichamrern S, Cheunsuchon B, Vasuvattakul S: Anti-CD20 Severity of primary MPGN, rather than MPGN type deter-
monoclonal antibody (rituximab) for the treatment of mines renal survival and post-transplant recurrence risk.
membranous nephropathy after living-unrelated kidney Kidney Int 69: 504 –511, 2006
transplantation: A case report. Transplant Proc 40: 2440 – 119. Morales JM: Hepatitis C virus infection and renal disease
2441, 2008 after renal transplantation. Transplant Proc 36: 760 –762,
105. El Kossi M, Harmer A, Goodwin J, Wagner B, Shortland J, 2004
Angel C, McKane W: De novo membranous nephropathy 120. Lien YH, Scott K: Long term cyclophosphamide treatment
associated with donor-specific alloantibody. Clin Trans- for recurrent type I membranoproliferative glomerulone-
plant 22: 124 –127, 2008 phritis after transplantation. Am J Kidney Dis 35: 539 –543,
106. Aline-Fardin A, Rifle G, Martin L, Justrabo E, Bour JB, 2000
D’Athis P, Tanter Y, Mousson C: Recurrent and de novo 121. Wu J, Jaar BG, Briggs WA, Choi MJ, Kraus ES, Racusen LC,
membranous glomerulopathy after kidney transplanta- Atta MG, Samaniego MD: High-dose mycophenolate
tion. Transplant Proc 41: 669 – 671, 2009 mofetil in the treatment of post-transplant glomerular dis-
107. El-Zoghby ZM, Grande JP, Fraile MG, Norby SM, Fervenza ease in the allograft: A case series. Nephron Clin Pract 98:
FC, Cosio FG: Recurrent idiopathic membranous nephrop- c61– 66, 2004
athy: Early diagnosis by protocol biopsies and treatment 122. Pickering MC, Cook HT: Translational mini-review se-
with anti-CD20 monoclonal antibodies. Am J Transplant 9: ries on complement factor H: Renal diseases associated
2800 –2807, 2009 with complement factor H: Novel insights form humans
108. Sprangers B, Lefkowitz GI, Cohen SD, Stokes MB, Valeri A, and animal. Clin Exp Immunol 151: 210 –230, 2008
Appel GB, Kunis CL: Beneficial effect of rituximab in the 123. Smith RJ, Alexander J, Barlow PN, Botto M, Cassavant TL,
treatment of recurrent idiopathic membranous nephropa- Cook HT, de Córdoba SR, Hageman GS, Jokiranta TS,
thy after kidney transplantation. Clin J Am Soc Nephrol 5: Kimberling WJ, Lambris JD, Lanning LD, Levidiotis V,
790 –797, 2010 Licht C, Lutz HU, Meri S, Pickering MC, Quigg RJ, Rops
109. Debiec H, Guigonis V, Mougenot B, Decobert F, Hay- AL, Salant DJ, Sethi S, Thurman JM, Tully HF, Tully SP,
mann JP, Bensman A, Deschênes G, Ronco PM: Antena- van der Vlag J, Walker PD, Würzner R, Zipfel PF; Dense
tal membranous glomerulonephritis due to anti-neutral Deposit Disease Focus Group: New approaches to the
2372 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 5: 2363–2372, 2010

treatment of dense deposit disease. J Am Soc Nephrol 18: mutation in the complement cofactor domain 15. Am J
2447–2456, 2007 Kidney Dis 45: 415– 421, 2005
124. Andresdottir MB: Recommendations for the diagnosis and 126. Saland JM, Ruggenenti P, Remuzzi G and the Consensus
treatment of dense deposit disease. Nat Clin Pract Nephrol 4: Study Group: Liver-kidney transplantation to cure atypical
68 – 69, 2008 hemolytic uremic syndrome. J Am Soc Nephrol 20: 940 –949,
125. Licht C, Weyersberg A, Heinen S, Stapenhorst L, Devenge 2009
J, Beck B, Waldherr R, Kirschfink M, Zipfel PF, Hoppe B: 127. Ponticelli C, Villa M, Cesana B, Montagnino G, Tarantino
Successful plasma therapy for atypical hemolytic uremic A: Risk factors for late kidney allograft failure. Kidney Int
syndrome caused by factor H deficiency owing to a novel 62: 1848 –1854, 2002

Potrebbero piacerti anche