You are on page 1of 11

Anti-Glomerular Basement Membrane Disease

Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

Stephen P. McAdoo and Charles D. Pusey

Abstract
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

Anti–glomerular basement membrane (anti-GBM) disease is a rare small vessel vasculitis that affects the capillary
beds of the kidneys and lungs. It is an archetypic autoimmune disease, caused by the development of directly
pathogenic autoantibodies targeting a well characterized autoantigen expressed in the basement membranes of
these organs, although the inciting events that induce the autoimmune response are not fully understood. The
Renal and Vascular
recent confirmation of spatial and temporal clustering of cases suggests that environmental factors, including Inflammation Section,
infection, may trigger disease in genetically susceptible individuals. The majority of patients develop widespread Department of
glomerular crescent formation, presenting with features of rapidly progressive GN, and 40%–60% will have Medicine, Imperial
concurrent alveolar hemorrhage. Treatment aims to rapidly remove pathogenic autoantibody, typically with the use College London,
London, United
of plasma exchange, along with steroids and cytotoxic therapy to prevent ongoing autoantibody production and
Kingdom
tissue inflammation. Retrospective cohort studies suggest that when this combination of treatment is started early,
the majority of patients will have good renal outcome, although presentation with oligoanuria, a high proportion of Correspondence: Dr.
glomerular crescents, or kidney failure requiring dialysis augur badly for renal prognosis. Relapse and recurrent Stephen P. McAdoo,
disease after kidney transplantation are both uncommon, although de novo anti-GBM disease after transplantation Renal and Vascular
for Alport syndrome is a recognized phenomenon. Copresentation with other kidney diseases such as ANCA- Inflammation Section,
Department of
associated vasculitis and membranous nephropathy seems to occur at a higher frequency than would be expected by
Medicine, Imperial
chance alone, and in addition atypical presentations of anti-GBM disease are increasingly reported. These observations College London,
highlight the need for future work to further delineate the immunopathogenic mechanisms of anti-GBM disease, and Hammersmith
how to better refine and improve treatments, particularly for patients presenting with adverse prognostic factors. Hospital Campus, Du
Cane Road, London
Clin J Am Soc Nephrol 12: 1162–1172, 2017. doi: https://doi.org/10.2215/CJN.01380217
W12 0NN. Email:
s.mcadoo@imperial.
ac.uk

Nomenclature and History techniques in the 1960s that it became possible to de-
Anti–glomerular basement membrane (anti-GBM) tect anti-GBM antibodies in kidney tissue (4), and to
disease is a rare small vessel vasculitis that affects demonstrate their pathogenic potential upon elution
glomerular capillaries (where it may result in glo- and transfer to nonhuman primates (5). The detection
merular necrosis and crescent formation), pulmonary of circulating anti-GBM antibodies in patients quickly
capillaries (where it may cause alveolar hemorrhage), followed (6), and the first comprehensive clinical de-
or both. It is characterized by the presence of circu- scription of “anti-GBM antibody–induced GN” was by
lating and deposited antibodies directed against Wilson and Dixon (7). It is of historical interest to note
basement membrane antigens, and as such is classified that Goodpasture’s original description of lung and
an immune-complex small vessel vasculitis in the kidney disease in association with intestinal and splenic
Revised International Chapel Hill Consensus Confer- inflammation, after a subacute clinical presentation, was
ence Nomenclature of Vasculitides (1). The Consensus perhaps more in keeping with a diagnosis of ANCA-
acknowledges the relative misnomer of anti-GBM associated vasculitis (AAV) than anti-GBM disease, and
disease, given the frequent involvement of alveolar that Goodpasture himself is said to have rejected the
basement membranes, although recognizes the widely eponymous use of his name.
accepted use of anti-GBM disease to describe this The term “Goodpasture disease” has persisted, how-
condition with or without lung involvement. ever, being generally reserved for patients with demon-
The eponymous term “Goodpasture disease” is also strable anti-GBM antibodies, whereas “Goodpasture
used to describe this condition, first being used by syndrome” may be used to describe copresentation
Australians Stanton and Tange in 1958 (2), in their with GN and pulmonary hemorrhage of any cause.
report describing nine cases of GN associated with We will use the term “anti-GBM GN” when referring
lung hemorrhage. They credited Ernest Goodpasture, specifically to the kidney involvement seen in this
an American pathologist, with the first description of condition, and “anti-GBM disease” when referring to
the syndrome in his 1919 paper describing a fatal case the broader spectrum of kidney and lung disease.
of GN and lung hemorrhage that was, at the time,
attributed to an atypic influenza infection (3). We do
not know, however, if any of these patients had anti- Epidemiology and Etiologic Associations
GBM disease as we recognize it today, because it was Given its rarity, definitive observations regarding
not until the development of immunofluorescence the incidence of anti-GBM disease are lacking. It is

1162 Copyright © 2017 by the American Society of Nephrology www.cjasn.org Vol 12 July, 2017
Clin J Am Soc Nephrol 12: 1162–1172, July, 2017 Anti-GBM disease, McAdoo et al. 1163

often said to have an incidence of ,1 per million population/yr populations (27,28). It should be noted, however, that these
in European populations, largely on the basis of single-center susceptibility alleles are common in most populations and
biopsy- or serology-based series, although accurately defining that they are also associated with other autoimmune
populations at risk in such studies is difficult. A recent study diseases (including multiple sclerosis, perhaps contributing
from Ireland is notable for being the first to define a nationwide to the association with alemtuzumab treatment), highlight-
disease incidence, by identifying all cases over a decade via ing that other factors are necessary to incite anti-GBM
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

reference immunology laboratories and a nationwide pathol- disease, and thus HLA-gene testing is not routinely used in
ogy database (8). It reported a disease rate of 1.64 per million the clinical work-up of these patients.
population/yr, higher than previous estimates. The disease is Polymorphisms and copy number variation in non-HLA
well recognized in other white and in Asian populations (9–12), genes have also been implicated in disease susceptibility,
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

although is thought to be rarer in African populations (13). such as the genes encoding Fcg-receptors (29,30), consis-
Anti-GBM GN accounts for 10%–15% of all cases of tent with the role of pathogenic autoantibodies in disease
crescentic GN in large biopsy series (14), although it ap- onset. On the basis of a small study, polymorphisms in
pears to be a rare cause of ESRD (15). A common observation COL4A3, the gene encoding the Goodpasture autoantigen,
from larger series of anti-GBM disease is that of a bimodal are not thought to be involved in disease predisposition
age distribution, with peak incidences in the third decade, (31). To the best of our knowledge, there has not been an
where a slight male preponderance and presentation with undirected genetic study in anti-GBM disease.
both kidney and lung disease is observed, and in the sixth to
seventh decades, where presentation with isolated kidney
disease is more common (16–18). Immunopathogenesis
Some series have reported disease “outbreaks” and In its native form, the GBM consists of a network of type
seasonal variation in incidence (16,17), and the Irish study IV collagen molecules, each made up of triple-helical
identified spatial and temporal clustering of disease, sug- protomers of a3, a4, and a5 chains (Figure 1). The principal
gesting that environmental factors may be important target of the autoimmune response in anti-GBM disease
triggers for disease onset, although they are yet to be has been identified as the noncollagenous (NC1) domain
accurately defined (8,19). Infectious associations, particu- of the a3 chain of type IV collagen (a3[IV]NC1; the
larly with influenza A, have been the subject of anecdotal “Goodpasture autoantigen”) (32,33). The clinical pattern of
reports (20,21), and may account for the aforementioned reno-pulmonary disease reflects the restricted expression of
seasonal or geographic “clustering” of anti-GBM disease this antigen to the basement membranes of glomerular and
cases, and a recent study described a high prevalence of alveolar capillaries (and to a lesser extent, the retina, choroid
prodromal upper and lower respiratory tract infection in a plexus, and cochlea, where it is generally not associated
cohort of 140 Chinese patients (22). The causative nature with clinical disease [34]). Two principle autoantibody (B cell)
of these associations, however, is not proven and remains epitopes within the autoantigen have been identified, desig-
speculative. nated EA and EB (35), which in native GBM are usually
A more conclusive environmental association is that with sequestered within the quaternary structure of the noncol-
cigarette smoking and the development of lung hemor- lagenous domains of the triple helix of a3, 4, and 5 chains.
rhage in anti-GBM disease (23). Similarly, inhalation of Sera from all patients with anti-GBM disease appear to
hydrocarbons has also been implicated in disease onset react to a3(IV)NC1, although a proportion will also have
(24). It is suggested that localized inflammation induced antibodies directed against other collagen chains, including
by inhaled toxins may increase capillary permeability, or a5 and a4, identified either in serum or upon elution from
potentially disrupt the quaternary structure of the alveolar kidney tissue, and thought to arise via a process of “epitope
basement membrane, exposing usually sequestered anti- spreading” after a primary response to the a3 chain (36).
gens and allowing access to pathogenic autoantibodies. The directly pathogenic potential of these antibodies was
A more recently identified trigger for anti-GBM disease clearly demonstrated by Lerner and colleagues in 1967,
is treatment with the anti-CD52 mAb, alemtuzumab, a when they administered antibodies eluted from the kid-
lymphocyte-depleting agent that is increasingly used in the neys of patients with anti-GBM disease to nonhuman
treatment of relapsing multiple sclerosis (25). It is thought primates, leading to the development of crescentic GN in
that loss of regulatory T cell subsets, or abnormal immune the recipients (5). The pathogenicity of these antibodies has
cell repopulation after depletion, may account for the in- since been confirmed in a number of other species and
creased incidence of many autoimmune diseases, including animal models.
anti-GBM disease, after exposure to this agent. Clinical observations support a pathogenic role for these
It is likely that these environmental triggers act in genet- antibodies; antibody titer, subclass, and avidity have each
ically susceptible individuals to induce disease onset. Anti- been correlated with disease outcome (37–40). In addition,
GBM disease has a strong HLA-gene association, with the rapid removal of circulating antibodies by plasma
approximately 80% of patients inheriting an HLA-DR2 exchange is associated with better outcome, and if kidney
haplotype. A hierarchy of associations with particular DRB1 transplantation is performed in the presence of circulating
alleles has been identified, some positively associated with antibodies, disease is likely to recur rapidly in the allograft
disease (DRB1*1501, DRB1*0401) and some conferring a (7,41).
dominant-negative protective effect (DRB1*07), which In addition to humoral responses, T cells also have a role
might be attributed to the higher affinity of the latter alleles in disease pathogenesis. Data from animal models sug-
for binding peptides from the target autoantigen (26). The gest that T cells may contribute directly to cell-mediated
DRB1*1501 association has been replicated in Asian glomerular injury, which can occur in the absence of
1164 Clinical Journal of the American Society of Nephrology
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

Figure 1. | Structure of the glomerular basement membrane. In its native form, the collagen IV network in the glomerular basement membrane
consists of triple-helical protomers of a3, a4, and a5 chains (shown individually in [A]). The carboxy-terminal domains of these a3 a4 a5
protomers form a trimeric “cap” (B), end-to-end association of which results in the formation of the hexameric NC1 domain (C). The quaternary
structure of this hexamer is stabilized by hydrophobic and hydrophilic interactions across the planar surfaces of opposing trimers, and reinforced
by sulfilimine bonds crosslinking opposing NC1 domains. Two key autoantibody epitopes within a3(IV)NC1 have been described, designated EA
(incorporating residues 17–31 toward the amino terminus) and EB (residues 127–141 toward the carboxy terminus), which in the native form are
sequestered at the junction with a4 and a5 chains within the triple helical structure. Binding through 7s domains (shown in orange) completes
the lattice-like structure of the type IV collagen network (D). Reprinted from reference 102.

significant humoral immunity (42,43), and glomerular T stabilize the association of opposing NC1 domains on
lymphocytes may be observed in kidney biopsy samples individual collagen chains (Figure 1). This may result in
taken from patients with active disease (44,45). The strong modification or exposure of usually hidden epitopes, which
HLA association and the presence of high-affinity, class- is suggested to be a key event in the pathogenesis of disease
switched autoantibodies also indicate a necessity for T cell (36). This may account for the association with etiologic
help in the development of the autoimmune response. factors that may disrupt alveolar (e.g., smoking, inhalation
Notably, mononuclear cells from patients proliferate in of hydrocarbons) or GBM (such as lithotripsy [51,52], and
response to a3(IV)NC1 at much higher frequency than do the other kidney pathologies discussed below).
cells from healthy controls, and the frequency of auto- The recovery phase of anti-GBM disease is associated
reactive T cells correlates with disease activity (46–48). The with a progressive fall in autoantibody titers (even in the
pathogenic T cell epitopes in humans, however, have not absence of immunosuppression) and a lower frequency of
been consistently defined. T cells reactive to a3(IV)NC1. The emergence of a CD251
That these autoreactive T cells can be identified in suppressor T cell subset that may inhibit responses to a3
healthy individuals, along with low-level natural autoan- (IV)NC1 has been described (53), suggesting that immu-
tibodies (49), suggests that tolerance to the a3(IV)NC1 nologic tolerance to a3(IV)NC1 can be re-established. This
antigen is not fully achieved during immunologic develop- may explain the rarity of clinical relapses in anti-GBM
ment. In addition, a rising titer of anti-GBM antibodies has disease, and the association with lymphocyte-depleting
been shown to predate the onset of clinical disease by therapy with alemtuzumab.
several months (50), highlighting that several tolerance
mechanisms must be disrupted before disease occurs. One
such breach of “peripheral” tolerance is disruption of the Clinical Presentation and Diagnosis
quaternary structure of the Goodpasture autoantigen, and The majority of patients (80%–90%) will present with
in particular disruption of the sulfilimine crosslinks that features of rapidly progressive GN. Forty percent to 60%
Clin J Am Soc Nephrol 12: 1162–1172, July, 2017 Anti-GBM disease, McAdoo et al. 1165

will have concurrent lung hemorrhage, and a small mi-


nority of patients may present with isolated pulmonary
disease. “Atypical” presentations are well recognized, and
discussed in more detail below. Central to the diagnosis of
anti-GBM disease is the identification of anti-GBM anti-
bodies, either in serum or deposited in tissue, along with
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

pathologic features of crescentic GN, with or without evi-


dence of alveolar hemorrhage.
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

Serologic Testing
In current practice, circulating anti-GBM antibodies are
usually detected using commercially available enzyme
immunoassays or bead-based fluorescence assays, which
typically use purified or recombinant human or animal
GBM preparations as antigenic substrate. Western blotting,
using similar GBM preparations, may be a more sensitive
method for antibody detection, although it is not Figure 2. | Kidney biopsy sample immunofluorescence for IgG re-
vealing linear deposits along the glomerular basement membrane,
widely available outside research laboratories. Indirect
and weaker staining of Bowman’s capsule and tubular basement
immunofluorescence using normal kidney tissue is an membranes.
alternative method, although this requires additional
input from a kidney pathologist and is prone to giving
false negative results. A proportion of patients who
have demonstrable deposition of IgG on the GBM by Conventional direct immunofluorescence techniques
immunofluorescence, but who are negative for circu- will identify all IgG subclasses, although will not differ-
lating antibodies by these conventional techniques, entiate the antigenic target of the kidney-bound antibody.
may be positive when tested by highly sensitive bio- Noncollagen chain antigens, such as entactin, have been
sensor assay (54). In anti-GBM disease, the pathogenic identified in historical case series, although their signifi-
antibodies are usually of the IgG class, with IgG1 and cance is not well characterized. In addition to detecting
IgG3 subclasses predominating (37,38), although rare deposited anti-GBM antibody, immunofluorescence may
cases of IgA- and IgG4-mediated disease have been demonstrate the presence of C components, in particular
described (55,56). These antibodies may not be detected C3 and C1q, along the GBM (17). A proportion of patients
on routine assays. may also demonstrate Igs or C deposition along tubular
Serologic testing for anti-GBM antibodies is, by defini- basement membranes.
tion, an urgent laboratory test, and we recommend that
results should be available within 24 hours for patients
presenting with RPGN, particularly when there are contra- Renal Biopsy Findings
Crescent formation is the histopathologic hallmark of
indications to kidney biopsy, because initiating treatment
anti-GBM disease (Figure 3). Large biopsy series suggest
before developing a need for RRT may have a significant
that 95% of patients will have evidence of crescent
effect on outcome. It should be noted, however, that
formation on kidney biopsy, and that in 80% of patients
approximately 10% of patients do not have identifiable
.50% of glomeruli will be affected. The average proportion
circulating antibodies with conventional assays, and so
of affected glomeruli is approximately 75% (14,57). The
serologic testing should not be the sole method of diagnosis
proportion of crescents observed in the biopsy sample
when kidney biopsy is available.
correlates strongly with the degree of renal impairment at
presentation (17,18). These crescents will typically be of
Deposited Antibody uniform age (Figure 3F), in contrast to other causes of
Direct immunofluorescence for Ig on frozen kidney RPGN, such as AAV, where a mixture of cellular, fibro-
tissue has high sensitivity for detecting deposited anti- cellular, and fibrous crescents may be seen. Crescentic
bodies, and is the gold-standard for diagnosis of anti-GBM glomeruli are likely to have areas of fibrinoid necrosis in the
disease, typically showing a strong linear ribbon-like underlying glomerular tuft. Noncrescentic glomeruli may
appearance (Figure 2). An important caveat is that fluo- similarly have segmental fibrinoid change (Figure 3A),
rescence may be negative or unclear in cases with severe although often they may appear completely normal. In
glomerular inflammation, where the underlying architec- early or mild disease, segmental proliferative change
ture is so disrupted that the linear pattern may not be may be seen, with infiltrating neutrophils or mononu-
recognized. Other causes of linear fluorescence should be clear lymphocytes. In severe disease, rupture of Bowman’s
considered (including diabetes, paraproteinemias, lupus capsule, peri-glomerular inflammation (Figure 3E), pro-
nephritis, and rarely fibrillary GN). Immunoperoxidase gressing to granuloma formation with multinucleate giant
techniques using paraffin-embedded tissue may also be cells, may be observed in a proportion. Given the acuity of
used, but may be less sensitive. Lung biopsy samples are disease onset, interstitial fibrosis and tubular atrophy are
not routinely used in the diagnosis of anti-GBM disease, uncommon in anti-GBM disease (unless there is preexisting
and, in our experience, immunofluorescence on lung tissue kidney pathology) although interstitial inflammation may
is rarely informative. be observed.
1166 Clinical Journal of the American Society of Nephrology
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

Figure 3. | Renal histopathology in anti–glomerular basement membrane (anti-GBM) GN. (A–C) Hematoxylin and eosin–stained sections
demonstrating (A) segmental fibrinoid necrotizing lesion in early anti-GBM GN; (B) small, circumscribed cellular crescent; and (C) large,
circumferential cellular crescent. (D–E) Demonstrate the use of Jones methylamine silver stain to delineate glomerular and tubular basement
membranes, clearly identifying a segmental area of extracapillary proliferation (D). (E) Demonstrates obliteration of the glomerular architecture
and rupture of Bowman’s capsule, with extravasation of red blood cells into the urinary space, and significant peri-glomerular inflammation.
(F) Adjacent glomeruli with synchronous cellular crescent formation typical of anti-GBM disease.

Electron microscopy is of limited additional value in the end-organ inflammation. The use of this combination of
diagnosis of anti-GBM disease, showing nonspecific fea- therapies was first described in 1976 (58), and they remain
tures of crescentic GN including rupture of the GBM and the core recommendation of the latest Kidney Disease
extracapillary localization of fibrin and proliferating cells. Improving Global Outcomes guideline for treating anti-
Electron-dense deposits are not seen in isolated anti-GBM GBM GN (59). We have reproduced a recommended
disease, although electron microscopy is necessary to ex- treatment schedule in Table 1 (13).
clude concomitant glomerular pathologies, such as mem- The inclusion of plasmapheresis is supported by obser-
branous GN, and may identify other diseases that may vational studies that suggest improved renal and patient
cause linear fluorescence (such as fibrillary GN and di- survival compared with historical cohorts treated with
abetic GBM thickening). immunosuppression alone (18,60). In addition, a large
contemporaneous Chinese study of 221 patients suggested
Diagnosis of Alveolar Hemorrhage better outcomes in patients who received plasmapheresis
Diffuse alveolar hemorrhage may be evident clinically, in addition to cytotoxic and corticosteroid therapy (61). To
or identified by radiologic examination. Broncho-alveolar date, there has only been one randomized trial in anti-GBM
lavage may identify hemosiderin-laden macrophages, a disease, which compared the addition of plasma exchange
characteristic feature of alveolar bleeding, and may also be to cyclophosphamide and steroids. Although this study
useful to exclude other pathologies, such as atypical in- was small (n517), the groups not ideally matched at
fection. In addition, pulmonary function testing, in partic- randomization, and its treatment regimens not represen-
ular the determination of the alveolar carbon monoxide tative of current practice, its findings supported the use of
transfer factor (KCO) may assist with the differentiation plasma exchange in anti-GBM disease (62). In particular, it
of alveolar hemorrhage from other causes of pulmonary demonstrated a much more rapid fall in circulating anti-
infiltration. The utility of both bronchoscopy and func- GBM antibodies and improved kidney function in patients
tional testing, however, may be limited by the clinical receiving plasmapheresis.
condition of the critically unwell patient. Immunoadsorption is an alternative form of extracor-
poreal therapy that may be more efficient than plasma
exchange for the removal of pathogenic autoantibody
Treatment (although conversely it may not remove proinflammatory
Standard treatment for anti-GBM disease includes plas- or procoagulant factors). In small series, it appears to have
mapheresis, to rapidly remove pathogenic autoantibody, comparable outcomes to plasma exchange therapy (63,64),
along with cyclophosphamide and corticosteroids, to in- and we note that a prospective, open-label study is planned
hibit further autoantibody production and to ameliorate to study the kinetics of anti-GBM antibody removal using
Clin J Am Soc Nephrol 12: 1162–1172, July, 2017 Anti-GBM disease, McAdoo et al. 1167

Table 1. Initial Treatment of Anti-GBM Disease

Agent Details and Duration Cautions

Plasma exchange Daily 4 L exchange for 5% human Monitor and correct as required: platelet
albumin solution. Add fresh human count, aim .70 3 109/L; fibrinogen,
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

plasma (300–600 ml) within 3 d of aim .1 g/L (may require


invasive procedure (e.g., kidney biopsy) cryoprecipitate supplementation to
or in patients with alveolar hemorrhage. support PEX); hemoglobin, aim for
Continue for 14 d or until antibody levels .90 g/L; corrected calcium, aim to
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

are fully suppressed. Monitor antibody keep in normal range


levels regularly after cessation of
treatment because plasma exchange may
require reinstatement if antibody levels
rebound.
Cyclophosphamide 2–3 mg/kg per d given orally for 2–3 mo. Stop if leukocyte count falls to ,4 3 109/
Reduce dose to 2 mg/kg in L and restart at reduced dose when
patients.55 yr. recovered. Insufficient evidence to
recommend use of IV
cyclophosphamide.
Corticosteroids Prednisolone 1 mg/kg per d (maximum 60 There is no evidence to support the use
mg) given orally. Reduce dose weekly to of methylprednisolone, and it may
20 mg by 6 wk, then gradually taper until increase the risk of infection
complete discontinuation at 6–9 mo.
Prophylactic treatments Prophylaxis against oropharyngeal fungal H2 receptor antagonists in those who are
infection (e.g., nystatin, amphotericin, or intolerant of PPI. Cotrimoxazole may
fluconazole) while on high-dose steroids. contribute to leukopenia; monitor
Peptic ulcer prophylaxis (e.g., with PPI) leukocyte count. Alternatives include
while on high-dose steroid treatment. nebulized pentamidine.
Prophylaxis against PCP (e.g.,
cotrimoxazole) while receiving high-
dose corticosteroids and
cyclophosphamide. Consider acyclovir
for CMV prophylaxis. Consider
prophylaxis against HBV reactivation
(e.g., lamivudine) in patients who have
evidence of previous infection (HBV cAb
positive).

Modified from reference 13, with permission. GBM, glomerular basement membrane; PEX, plasma exchange; IV, intravenous; PPI,
proton pump inhibitor; PCP, Pneumocystis jiroveci pneumonia; CMV, cytomegalovirus; HBV, hepatitis B virus; cAb, core antibody.

this technique (NCT02765789), which may be considered an cases or small series (67–69). There is insufficient evidence,
alternative depending on local availability. at present, to recommend their use in first-line therapy,
In AAV, the equivalence of daily oral and pulsed intra- although they may be considered in patients who have
venous cyclophosphamide in induction therapy has been contra-indication or intolerance to conventional treatment.
established in a large randomized controlled trial (65). In addition to targeted immunotherapy, patients may
Nearly all published experience in anti-GBM GN, however, require immediate organ support; in larger series, approx-
has used daily oral dosing, and so we recommend this as imately half of patients require hemodialysis at the point of
the first-line approach in this disease. Because the risk of initial presentation (18). There are limited data on how
relapse is very low, and approximately only 3 months of frequently artificial ventilation is required, although one
cytotoxic therapy is usually required, concerns about total small series estimated that this occurred in 11% of patients
cumulative dose of cyclophosphamide are less relevant with lung hemorrhage (70). There are case reports of
than in AAV. In our experience, high-dose intravenous gluco- successful use of extracorporeal membrane oxygenation in
corticoids are not required in the treatment of anti-GBM patients with very severe lung disease (71–73).
disease, provided the other components of therapy, in
particular plasma exchange, can be initiated promptly (18).
The use of other immunosuppressive therapies in anti- Outcome and Prognosis
GBM disease is less well described. There are several Long-term follow-up of the largest cohort of patients
reports of rituximab use, as either “add-on” to standard (n571) all treated with the combination of plasma ex-
therapy or as a substitute for cyclophosphamide in patients change, cyclophosphamide, and corticosteroids, from the
who are intolerant (66). Similarly, the use of mycophenolate Hammersmith Hospital, London, United Kingdom, sug-
mofetil and cyclosporine has been reported in individual gests that it is effective in treating lung hemorrhage in
1168 Clinical Journal of the American Society of Nephrology

.90%, and in preserving independent kidney function in recurrence in the allograft, at frequencies of up to 50%
the majority of patients, including those who present in historical series (41). Most centers therefore recommend
with severe kidney dysfunction (18). In patients presenting a period of at least 6 months’ sustained seronegativ-
with creatinine values ,500 mmol/L, renal survival was ity before undertaking transplantation in patients who
95% and 94% at 1 and 5 years, respectively. In patients have reached ESRD due to anti-GBM disease (59). Under
presenting with creatinine .500 mmol/L, but not requiring these circumstances, and with current immunosuppres-
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

immediate dialysis, renal survival was 82% and 50% at sive regimens, recurrent disease is rare; the ANZDATA
the same respective time-points. In patients presenting registry study found that 6 of 449 (2.7%) patients de-
with an initial requirement for dialysis, however, renal veloped biopsy-proven recurrent anti-GBM disease,
recovery occurred in only 8% at 1 year. Other reports have which led to graft failure in two cases (15). The frequency
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

described similarly low levels of renal recovery in patients of other causes of graft failure was similar to patients
presenting with dialysis-dependent kidney failure, with the transplanted for ESRD of other causes, and overall patient
highest rate of approximately 20% recovery in one series (66). and renal survival in anti-GBM disease was similar to
Predictors of poor renal outcome include severity of other groups in this study. These findings were somewhat
renal dysfunction at presentation, the proportion of glomer- in contrast to a previous European study that suggested
uli affected by crescents, and oligoanuria at presenta- patient survival was favorable in patients transplanted for
tion (17,18,74). In the Hammersmith series, no patient who anti-GBM disease compared with those transplanted for
required hemodialysis and had 100% crescents on kidney other primary kidney diseases (although significant dif-
biopsy recovered renal function, and so withholding treat- ferences in age at transplantation may account for this
ment (and its incumbent toxicity) is often considered in these apparent difference in patient survival) (78). The Euro-
cases. An isolated case of renal recovery despite these pean study also reported a higher frequency of recurrent
findings (75), however, highlights the need to consider all disease (14%), although this may reflect differences in
cases for treatment, with specific attention to other features immunosuppressive use during an earlier era, and the
that might predict renal recovery on biopsy (such as study did not comment on anti-GBM titers and their
concomitant acute tubular injury) and the ability of patients relationship to timing of transplantation. In the current
to tolerate each component of therapy. A short trial of early era, isolated case reports of recurrent disease still exist
treatment may be considered, and rapidly tapered if there is (79).
no evidence of renal recovery within 2–4 weeks. In addition,
the potential benefit of a period of immunosuppression to
expedite autoantibody clearance, thus allowing earlier kid- Post-Transplant Anti-GBM Disease in Alport
ney transplantation, should be considered in suitable pa- Syndrome
tients. We have used rituximab monotherapy, for example, Mutations in any of the genes which encode the a3, a4, or
in patients with ESRD who have an identified live-donor a5 chains may result in a failure to produce the normal type
for transplantation, but remain anti-GBM antibody–positive, IV collagen network present in GBM, and thus lead to
although controlled evidence for this indication is lacking. progressive kidney disease in Alport Syndrome. Mutations
A recent retrospective study (an Australia and New in the COL4A5 gene located on the X chromosome are most
Zealand Dialysis and Transplant Registry [ANZDATA] common, giving rise to typical X-linked Alport syndrome,
study) analyzed the long-term outcomes of 449 patients although autosomal recessive and dominant disease are
with ESRD due to anti-GBM disease, and found that their recognized with COL4A3 and COL4A4 mutations. After
survival was comparable to patients with ESRD of other kidney transplantation, these patients may develop anti-
causes, whether they remained on dialysis or underwent GBM antibodies as an alloimmune response to the neo-
kidney transplantation (15). Chronic respiratory sequelae antigens contained in “normal” a3, a4, or a5 chains in the
after alveolar hemorrhage are uncommon (70). kidney allograft. In X-linked disease, these antibodies do
Relapse is rare in anti-GBM disease, occurring in ,3% of not recognize the individual EA and EB epitopes of the a3
patients in the Hammersmith series (18). It is usually as- chain recognized by sera from Goodpasture patients, but
sociated with ongoing exposure to pulmonary irritants rather a distinct, composite epitope on the a5 chain, that is
such as cigarette smoke and hydrocarbons (76,77), and not sequestered within the native hexamer of the Goopas-
avoidance of these precipitants is an essential part of ture antigen (36). It should be noted that commercially
long-term management of these cases. We recommend available anti-GBM assays, which are optimized to detect
repeat kidney biopsy in cases of relapse with kidney reactivity to the a3(IV)NC1 antigen, may fail to detect
involvement, in order to secure an accurate diagnosis and circulating antibodies in this setting. Anti-GBM anti-
to exclude concomitant pathologies such as AAV and bodies may be detected in 5%–10% of Alport patients
membranous nephropathy (discussed below). In confirmed after transplantation, although the development of overt
cases, standard retreatment with cytotoxics and corticoste- GN in the allograft is less frequent (perhaps owing to the
roids is usually indicated. In a patient with multiply effects of maintenance immunosuppression). When GN
relapsing alveolar hemorrhage we have found treatment develops, however, it usually occurs early and carries a high
with rituximab beneficial. risk of graft loss (80,81). Repeated transplantation in this
setting almost invariably leads to more aggressive disease
recurrence and rapid graft loss, and is undertaken at very
Kidney Transplantation after Anti-GBM Disease high risk (82). Individuals with large COL4A5 gene dele-
Kidney transplantation performed in the presence of tions are at increased risk of post-transplant anti-GBM
anti-GBM antibodies results in a high likelihood of disease disease, and recent guidelines encourage the use of genetic
Clin J Am Soc Nephrol 12: 1162–1172, July, 2017 Anti-GBM disease, McAdoo et al. 1169

testing to inform discussions regarding the risk of de novo during splenectomy for hypersplenism and a diag-
anti-GBM disease after transplantation (83). nostic workup for optic vasculitis, who was treated with
steroids only, and who had normal renal function after
1 year follow-up (7). There have been a number of series of
Other Variant Forms of Anti-GBM Disease “atypical” cases published in recent years, often with less
Double-Positive Anti-GBM and ANCA-Associated GN severe renal involvement than is seen in the classic pre-
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

The concurrence of ANCA and anti-GBM antibodies is sentation of anti-GBM disease, although it not always clear
recognized to occur at much higher frequency than ex- whether these represent distinct clinical subphenotypes or
pected by chance alone. In some series, almost half of heterogenous cases on a spectrum of disease severity (92–95).
The largest of these series, reported by Nasr and
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

patients with anti-GBM disease have detectable ANCA


(usually recognizing myeloperoxidase [MPO]), and up to colleagues, described 20 patients with mild and indolently
10% of patients with ANCA also have circulating anti-GBM progressive renal impairment, who had linear Ig deposi-
antibodies (84–86). The mechanism of this association is tion on kidney biopsy, but without predominant features of
unclear, although it has been shown that ANCA may be crescentic GN, and without overt lung hemorrhage (95).
detected before the onset of anti-GBM disease, suggesting Circulating anti-GBM antibodies were not detected using
that ANCA-induced glomerular inflammation may be a conventional assays, and both patient and renal prognosis
trigger for the development of an anti-GBM response, were good, with 90% and 85% survival at 1 year, re-
perhaps by modifying or exposing usually sequestered spectively. They estimated that these atypical cases ac-
disease epitopes in GBM (50). Conversely, a recent study counted for approximately 10% of anti-GBM cases at their
found that up to 60% of anti-GBM cases also had anti- center. Notably, half of the cases had light-chain restric-
bodies directed against linear epitopes of MPO, versus 24% tion on immunofluorescence, although the authors suggest
recognizing intact MPO. The authors hypothesize that that the pathologic features were not in keeping with
MPO-ANCA recognizing linear and conformational epi- proliferative GN with monoclonal Ig deposition. They
topes may arise sequentially, via a process of inter- and suggest that differences in the antigen specificity, Ig sub-
intramolecular epitope spreading (87). We recently ana- class, and/or the ability to fix C and recruit inflammatory
lyzed the outcomes of a large cohort of these “double- cells, of these atyptical compared with “classic” anti-GBM an-
positive” patients from four centers in Europe, and found tibodies, account for the less severe disease phenotype seen.
that they experience the early morbidity and mortality of Another small but well characterized series with a
anti-GBM disease, with severe kidney and lung disease at distinct clinical phenotype was recently described in
presentation, requiring aggressive immunosuppres- Sweden; it included four young females, who had severe
sive therapy and plasma exchange (88). During long-term lung disease and minimal kidney involvement, who were
follow-up, they relapsed at a frequency comparable to a found to have IgG4 subclass anti-GBM antibodies that were
parallel cohort of patients with AAV, suggesting they not detectable with conventional anti-GBM assays (56). That
warrant more careful long-term follow-up and maintenance two of these patients demonstrated higher signal in the anti-
immunosuppression, unlike patients with single-positive GBM ELISA when using a nondenaturing buffer suggests
anti-GBM disease. that differences in epitope specificity might also account for
the negative testing seen with the routine assays, and
supports the hypothesis that differences in clinical pre-
Anti-GBM Disease Associated with Membranous sentation might be related to differences in the subclass or
Nephropathy target of the anti-GBM antibody.
There are several reports of anti-GBM disease associated
with membranous nephropathy, occurring as a preceding,
simultaneous, or succeeding diagnosis (89,90). As with
the ANCA association, it is postulated that disruption of
Future Directions
Despite being one of the better characterized autoim-
glomerular architecture by one disease reveals hidden
mune diseases, unanswered questions remain regarding
epitopes that allow the second process to occur. A rapid
the pathogenesis of anti-GBM disease, which may have
decline in kidney function in a patient with known
important clinical implications. These include the need to
membranous nephropathy should raise suspicion of the
further characterize the variant forms of disease, and how
development of superimposed crescentic nephritis or anti-
differences in antibody subclass or specificity might influ-
GBM disease, and rebiopsy is recommended. We suggest
ence presentation, the appropriate use of treatment, and
that these cases are treated initially as for anti-GBM disease,
outcomes. Better understanding of T cell functions, and in
although how they should be managed in the long-term is
particular the role of regulatory cells that may suppress
not clear. The authors of a recent case report suggest that
disease, may have therapeutic significance, both in anti-
rituximab may be a useful agent to treat both pathologies
GBM disease and other autoimmune conditions. The
simultaneously (91).
induction of immunologic tolerance using mucosal admin-
istration of GBM antigen has been described in experimental
“Atypical” Anti-GBM Disease models (96), which may likewise have therapeutic potential.
Unusual presentations of anti-GBM disease have been Finally, the inciting events that cause autoimmunity to GBM
recognized for as long as the disease itself. Wilson and antigens remain unclear. Idiotype–anti-idiotype interactions
Dixon’s original 1973 report, for example, included the have been invoked in a recent study (97), and the role of
case of a male 14-year-old who had the incidental finding infectious triggers that might operate via a similar mechanism
of linear IgG deposition on a kidney biopsy sample taken in clinical disease induction could be explored further.
1170 Clinical Journal of the American Society of Nephrology

As a rare disorder requiring immediate treatment, co- 7. Wilson CB, Dixon FJ: Anti-glomerular basement membrane
ordinating large, prospective studies in anti-GBM disease is antibody-induced glomerulonephritis. Kidney Int 3: 74–89,
1973
challenging. In addition, the efficacy of current treatment 8. Canney M, O’Hara PV, McEvoy CM, Medani S, Connaughton
regimens, when started early enough, is widely accepted. DM, Abdalla AA, Doyle R, Stack AG, O’Seaghdha CM, Clarkson
Future therapeutic studies, therefore, should perhaps focus on MR, Griffin MD, Holian J, Dorman AM, Niland A, Keogan M,
identifying “add-on” treatments that might improve outcomes Wallace EM, Conlon NP, Walsh C, Kelly A, Little MA: Spatial and
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

in severe disease. We have recently shown that treatment with temporal clustering of anti-glomerular basement membrane
disease. Clin J Am Soc Nephrol 11: 1392–1399, 2016
fostamatinib, a spleen tyrosine kinase (SYK) inhibitor, effec- 9. McPhaul JJ Jr, Mullins JD: Glomerulonephritis mediated by
tively reverses crescent formation in rodent models of anti- antibody to glomerular basement membrane. Immunological,
GBM disease (98,99) (and that intraglomerular SYK can be clinical, and histopathological characteristics. J Clin Invest 57:
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

detected in patient kidney biopsy samples [100]), so it would 351–361, 1976


10. Taylor DM, Yehia M, Simpson IJ, Thein H, Chang Y, de Zoysa JR:
be of interest to explore the use of this agent in advanced
Anti-glomerular basement membrane disease in Auckland.
clinical disease. Another interesting agent that has shown Intern Med J 42: 672–676, 2012
efficacy in experimental models is IdeS (IgG-degrading en- 11. Hirayama K, Yamagata K, Kobayashi M, Koyama A: Anti-glo-
zyme of Streptococcus pyogenes), an enzyme that is able to merular basement membrane antibody disease in Japan: Part of
cleave both circulating and membrane-bound Ig (101). IdeS the nationwide rapidly progressive glomerulonephritis survey in
Japan. Clin Exp Nephrol 12: 339–347, 2008
was safe and tolerable in early-phase human studies, and a 12. Li FK, Tse KC, Lam MF, Yip TP, Lui SL, Chan GS, Chan KW, Chan
clinical study in severe anti-GBM disease, where it may EY, Choy BY, Lo WK, Chan TM, Lai KN: Incidence and outcome
promote rapid clearance of pathogenic IgG, has been proposed of antiglomerular basement membrane disease in Chinese.
(EudraCT number: 2016–004082–39). Finally, large multicenter Nephrology (Carlton) 9: 100–104, 2004
13. Pusey CD: Anti-glomerular basement membrane disease. Kid-
studies might aim to identify clinical and histopatho-
ney Int 64: 1535–1550, 2003
logic indicators that reliably predict failure to respond to 14. Jennette JC: Rapidly progressive crescentic glomerulonephritis.
treatment, so that the toxicities associated with intensive Kidney Int 63: 1164–1177, 2003
immunosuppression may be avoided in futile cases. 15. Tang W, McDonald SP, Hawley CM, Badve SV, Boudville NC,
As an archetypal autoimmune disorder, such studies of Brown FG, Clayton PA, Campbell SB, de Zoysa JR, Johnson DW:
Anti-glomerular basement membrane antibody disease is an
anti-GBM disease and its experimental correlates are likely uncommon cause of end-stage renal disease. Kidney Int 83: 503–
to provide fresh insights into the mechanisms of renal 510, 2013
autoimmunity. However, it is the alarming clinical pre- 16. Savage CO, Pusey CD, Bowman C, Rees AJ, Lockwood CM:
sentation, and the need for emergency treatment, often in Antiglomerular basement membrane antibody mediated dis-
ease in the British Isles 1980-4. Br Med J (Clin Res Ed) 292: 301–
critically unwell patients, that underscores the need for
304, 1986
clinicians to be mindful of this rare condition, the pitfalls 17. Fischer EG, Lager DJ: Anti-glomerular basement membrane
associated with its diagnosis, particularly in atypical and glomerulonephritis: A morphologic study of 80 cases. Am J Clin
variant presentations, and the early and appropriate use of Pathol 125: 445–450, 2006
immunosuppressive and extracorporeal therapies, in order 18. Levy JB, Turner AN, Rees AJ, Pusey CD: Long-term outcome of
anti-glomerular basement membrane antibody disease treated
to prevent morbidity and to improve survival. with plasma exchange and immunosuppression. Ann Intern
Med 134: 1033–1042, 2001
Acknowledgments 19. McAdoo SP, Pusey CD: Clustering of Anti-GBM Disease: Clues
We acknowledge support from the National Institute for Health to an environmental trigger? Clin J Am Soc Nephrol 11: 1324–
Research Imperial Biomedical Research Centre. 1326, 2016
20. Perez GO, Bjornsson S, Ross AH, Aamato J, Rothfield N: A mini-
Disclosures epidemic of Goodpasture’s syndrome clinical and immuno-
logical studies. Nephron 13: 161–173, 1974
None.
21. Wilson CB, Smith RC: Goodpasture’s syndrome associated with
influenza A2 virus infection. Ann Intern Med 76: 91–94, 1972
References 22. Gu QH, Xie LJ, Jia XY, Ma R, Liao YH, Cui Z, Zhao MH: Fever
1. Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, Flores- and prodromal infections in anti-glomerular basement mem-
Suarez LF, Gross WL, Guillevin L, Hagen EC, Hoffman GS, Jayne brane disease [published online ahead of print March 6, 2017].
DR, Kallenberg CG, Lamprecht P, Langford CA, Luqmani RA, Nephrology (Carlton) doi:10.1111/nep.13040
Mahr AD, Matteson EL, Merkel PA, Ozen S, Pusey CD, 23. Donaghy M, Rees AJ: Cigarette smoking and lung haemorrhage
Rasmussen N, Rees AJ, Scott DG, Specks U, Stone JH, Takahashi in glomerulonephritis caused by autoantibodies to glomerular
K, Watts RA: 2012 revised International Chapel Hill Consensus basement membrane. Lancet 2: 1390–1393, 1983
Conference Nomenclature of Vasculitides. Arthritis Rheum 65: 24. Bombassei GJ, Kaplan AA: The association between hydrocar-
1–11, 2013 bon exposure and anti-glomerular basement membrane
2. Stanton MC, Tange JD: Goodpasture’s syndrome (pulmonary antibody-mediated disease (Goodpasture’s syndrome).
haemorrhage associated with glomerulonephritis). Australas Am J Ind Med 21: 141–153, 1992
Ann Med 7: 132–144, 1958 25. Clatworthy MR, Wallin EF, Jayne DR: Anti-glomerular basement
3. Goodpasture E: The significance of certain pulmonary lesions in re- membrane disease after alemtuzumab. N Engl J Med 359: 768–
lation to the etiology of influenza. Am J Med Sci 158: 863–870, 1919 769, 2008
4. Scheer RL, Grossman MA: Immune aspects of the glomerulo- 26. Fisher M, Pusey CD, Vaughan RW, Rees AJ: Susceptibility to
nephritis associated with pulmonary hemorrhage. Ann Intern anti-glomerular basement membrane disease is strongly asso-
Med 60: 1009–1021, 1964 ciated with HLA-DRB1 genes. Kidney Int 51: 222–229, 1997
5. Lerner RA, Glassock RJ, Dixon FJ: The role of anti-glomerular 27. Yang R, Cui Z, Zhao J, Zhao MH: The role of HLA-DRB1 alleles on
basement membrane antibody in the pathogenesis of human susceptibility of Chinese patients with anti-GBM disease. Clin
glomerulonephritis. J Exp Med 126: 989–1004, 1967 Immunol 133: 245–250, 2009
6. McPhaul JJ Jr, Dixon FJ: The presence of anti-glomerular base- 28. Kitagawa W, Imai H, Komatsuda A, Maki N, Wakui H, Hiki Y,
ment membrane antibodies in peripheral blood. J Immunol 103: Sugiyama S: The HLA-DRB1*1501 allele is prevalent among
1168–1175, 1969 Japanese patients with anti-glomerular basement membrane
Clin J Am Soc Nephrol 12: 1162–1172, July, 2017 Anti-GBM disease, McAdoo et al. 1171

antibody-mediated disease. Nephrol Dial Transplant 23: 3126– 49. Cui Z, Zhao MH, Segelmark M, Hellmark T: Natural autoanti-
3129, 2008 bodies to myeloperoxidase, proteinase 3, and the glomerular
29. Zhou XJ, Lv JC, Bu DF, Yu L, Yang YR, Zhao J, Cui Z, Yang R, Zhao basement membrane are present in normal individuals. Kidney
MH, Zhang H: Copy number variation of FCGR3A rather than Int 78: 590–597, 2010
FCGR3B and FCGR2B is associated with susceptibility to anti- 50. Olson SW, Arbogast CB, Baker TP, Owshalimpur D, Oliver DK,
GBM disease. Int Immunol 22: 45–51, 2010 Abbott KC, Yuan CM: Asymptomatic autoantibodies associate
30. Zhou XJ, Lv JC, Yu L, Cui Z, Zhao J, Yang R, Han J, Hou P, Zhao with future anti-glomerular basement membrane disease. J Am
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

MH, Zhang H: FCGR2B gene polymorphism rather than Soc Nephrol 22: 1946–1952, 2011
FCGR2A, FCGR3A and FCGR3B is associated with anti-GBM 51. Xenocostas A, Jothy S, Collins B, Loertscher R, Levy M: Anti-
disease in Chinese. Nephrol Dial Transplant 25: 97–101, 2010 glomerular basement membrane glomerulonephritis after
31. Persson U, Hertz JM, Carlsson M, Hellmark T, Juncker I, extracorporeal shock wave lithotripsy. Am J Kidney Dis 33: 128–
Wieslander J, Segelmark M: Patients with Goodpasture’s disease 132, 1999
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

have two normal COL4A3 alleles encoding the NC1 domain of 52. Guerin V, Rabian C, Noel LH, Droz D, Baron C, Lallemand F,
the type IV collagen alpha 3 chain. Nephrol Dial Transplant 19: Jungers P: Anti-glomerular-basement-membrane disease after
2030–2035, 2004 lithotripsy. Lancet 335: 856–857, 1990
32. Saus J, Wieslander J, Langeveld JP, Quinones S, Hudson BG: 53. Salama AD, Chaudhry AN, Holthaus KA, Mosley K, Kalluri R,
Identification of the Goodpasture antigen as the alpha 3(IV) Sayegh MH, Lechler RI, Pusey CD, Lightstone L: Regulation by
chain of collagen IV. J Biol Chem 263: 13374–13380, 1988 CD251 lymphocytes of autoantigen-specific T-cell responses in
33. Turner N, Mason PJ, Brown R, Fox M, Povey S, Rees A, Pusey CD: Goodpasture’s (anti-GBM) disease. Kidney Int 64: 1685–1694,
Molecular cloning of the human Goodpasture antigen demon- 2003
strates it to be the alpha 3 chain of type IV collagen. J Clin Invest 54. Salama AD, Dougan T, Levy JB, Cook HT, Morgan SH, Naudeer S,
89: 592–601, 1992 Maidment G, George AJ, Evans D, Lightstone L, Pusey CD:
34. Cashman SJ, Pusey CD, Evans DJ: Extraglomerular distribution of Goodpasture’s disease in the absence of circulating anti-
immunoreactive Goodpasture antigen. J Pathol 155: 61–70, 1988 glomerular basement membrane antibodies as detected by
35. Netzer KO, Leinonen A, Boutaud A, Borza DB, Todd P, Gunwar standard techniques. Am J Kidney Dis 39: 1162–1167, 2002
S, Langeveld JP, Hudson BG: The goodpasture autoantigen. 55. Moulis G, Huart A, Guitard J, Fortenfant F, Chauveau D: IgA-
Mapping the major conformational epitope(s) of alpha3(IV) mediated anti-glomerular basement membrane disease: An
collagen to residues 17-31 and 127-141 of the NC1 domain. uncommon mechanism of Goodpasture’s syndrome. Clin Kid-
J Biol Chem 274: 11267–11274, 1999 ney J 5: 545–548, 2012
36. Pedchenko V, Bondar O, Fogo AB, Vanacore R, Voziyan P, 56. Ohlsson S, Herlitz H, Lundberg S, Selga D, Mölne J, Wieslander J,
Kitching AR, Wieslander J, Kashtan C, Borza DB, Neilson EG, Segelmark M: Circulating anti-glomerular basement membrane
Wilson CB, Hudson BG: Molecular architecture of the Good- antibodies with predominance of subclass IgG4 and false-
pasture autoantigen in anti-GBM nephritis. N Engl J Med 363: negative immunoassay test results in anti-glomerular basement
343–354, 2010 membrane disease. Am J Kidney Dis 63: 289–293, 2014
37. Zhao J, Yan Y, Cui Z, Yang R, Zhao MH: The immunoglobulin G 57. Jennette JC, Thomas DB: Crescentic glomerulonephritis.
subclass distribution of anti-GBM autoantibodies against Nephrol Dial Transplant 16[Suppl 6]: 80–82, 2001
rHalpha3(IV)NC1 is associated with disease severity. Hum Im- 58. Lockwood CM, Rees AJ, Pearson TA, Evans DJ, Peters DK, Wilson
munol 70: 425–429, 2009 CB: Immunosuppression and plasma-exchange in the treatment
38. Segelmark M, Butkowski R, Wieslander J: Antigen restriction and of Goodpasture’s syndrome. Lancet 1: 711–715, 1976
IgG subclasses among anti-GBM autoantibodies. Nephrol Dial 59. Kidney Disease Improving Global Outcomes (KDIGO) Glo-
Transplant 5: 991–996, 1990 merulonephritis Work Group: KDIGO clinical practice
39. Cui Z, Zhao MH: Avidity of anti-glomerular basement mem- guideline for glomerulonephritis. Kidney Int Suppl 2: 139–274,
brane autoantibodies was associated with disease severity. Clin 2012
Immunol 116: 77–82, 2005 60. Simpson IJ, Doak PB, Williams LC, Blacklock HA, Hill RS,
40. Hellmark T, Segelmark M, Unger C, Burkhardt H, Saus J, Teague CA, Herdson PB, Wilson CB: Plasma exchange in
Wieslander J: Identification of a clinically relevant im- Goodpasture’s syndrome. Am J Nephrol 2: 301–311, 1982
munodominant region of collagen IV in Goodpasture disease. 61. Cui Z, Zhao J, Jia XY, Zhu SN, Jin QZ, Cheng XY, Zhao MH: Anti-
Kidney Int 55: 936–944, 1999 glomerular basement membrane disease: Outcomes of different
41. Choy BY, Chan TM, Lai KN: Recurrent glomerulonephritis after therapeutic regimens in a large single-center Chinese cohort
kidney transplantation. Am J Transplant 6: 2535–2542, 2006 study. Medicine (Baltimore) 90: 303–311, 2011
42. Dean EG, Wilson GR, Li M, Edgtton KL, O’Sullivan KM, Hudson 62. Johnson JP, Moore J Jr, Austin HA 3rd, Balow JE, Antonovych TT,
BG, Holdsworth SR, Kitching AR: Experimental autoimmune Wilson CB: Therapy of anti-glomerular basement membrane
Goodpasture’s disease: A pathogenetic role for both effector antibody disease: Analysis of prognostic significance of clinical,
cells and antibody in injury. Kidney Int 67: 566–575, 2005 pathologic and treatment factors. Medicine (Baltimore) 64: 219–
43. Wu J, Hicks J, Borillo J, Glass WF 2nd, Lou YH: CD4(1) T cells 227, 1985
specific to a glomerular basement membrane antigen mediate 63. Biesenbach P, Kain R, Derfler K, Perkmann T, Soleiman A,
glomerulonephritis. J Clin Invest 109: 517–524, 2002 Benharkou A, Druml W, Rees A, Säemann MD: Long-term
44. Bolton WK, Innes DJ Jr, Sturgill BC, Kaiser DL: T-cells and outcome of anti-glomerular basement membrane antibody
macrophages in rapidly progressive glomerulonephritis: Clini- disease treated with immunoadsorption. PLoS One 9: e103568,
copathologic correlations. Kidney Int 32: 869–876, 1987 2014
45. Nolasco FE, Cameron JS, Hartley B, Coelho A, Hildreth G, 64. Zhang YY, Tang Z, Chen DM, Gong DH, Ji DX, Liu ZH: Com-
Reuben R: Intraglomerular T cells and monocytes in nephritis: parison of double filtration plasmapheresis with immuno-
Study with monoclonal antibodies. Kidney Int 31: 1160–1166, adsorption therapy in patients with anti-glomerular basement
1987 membrane nephritis. BMC Nephrol 15: 128, 2014
46. Derry CJ, Ross CN, Lombardi G, Mason PD, Rees AJ, Lechler RI, 65. de Groot K, Harper L, Jayne DR, Flores Suarez LF, Gregorini G,
Pusey CD: Analysis of T cell responses to the autoantigen Gross WL, Luqmani R, Pusey CD, Rasmussen N, Sinico RA, Tesar
in Goodpasture’s disease. Clin Exp Immunol 100: 262–268, V, Vanhille P, Westman K, Savage CO; EUVAS (European
1995 Vasculitis Study Group): Pulse versus daily oral cyclophospha-
47. Zou J, Hannier S, Cairns LS, Barker RN, Rees AJ, Turner AN, mide for induction of remission in antineutrophil cytoplasmic
Phelps RG: Healthy individuals have Goodpasture autoantigen- antibody-associated vasculitis: A randomized trial. Ann Intern
reactive T cells. J Am Soc Nephrol 19: 396–404, 2008 Med 150: 670–680, 2009
48. Salama AD, Chaudhry AN, Ryan JJ, Eren E, Levy JB, Pusey CD, 66. Touzot M, Poisson J, Faguer S, Ribes D, Cohen P, Geffray L,
Lightstone L, Lechler RI: In Goodpasture’s disease, CD4(1) T Anguel N, François H, Karras A, Cacoub P, Durrbach A, Saadoun
cells escape thymic deletion and are reactive with the autoan- D: Rituximab in anti-GBM disease: A retrospective study of 8
tigen alpha3(IV)NC1. J Am Soc Nephrol 12: 1908–1915, 2001 patients. J Autoimmun 60: 74–79, 2015
1172 Clinical Journal of the American Society of Nephrology

67. Mori M, Nwaogwugwu U, Akers GR, McGill RL: Anti- membrane antibodies and myeloperoxidase-ANCAs in cres-
glomerular basement membrane disease treated with centic glomerulonephritis. Am J Kidney Dis 46: 253–262, 2005
mycophenolate mofetil, corticosteroids, and plasmapheresis. 87. Li JN, Cui Z, Wang J, Hu SY, Jia XY, Guan Z, Chen M, Xie C, Zhao
Clin Nephrol 80: 67–71, 2013 MH: Autoantibodies against linear epitopes of myeloperoxidase
68. Garcı́a-Cantón C, Toledo A, Palomar R, Fernandez F, Lopez J, in anti-glomerular basement membrane disease. Clin J Am Soc
Moreno A, Esparza N, Suria S, Rossique P, Diaz JM, Checa D: Nephrol 11: 568–575, 2016
Goodpasture’s syndrome treated with mycophenolate mofetil. 88. McAdoo SP, Tanna A, Hrušková Z, Holm L, Weiner M,
Downloaded from http://journals.lww.com/cjasn by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1A

Nephrol Dial Transplant 15: 920–922, 2000 Arulkumaran N, Kang A, Satrapová V, Levy J, Ohlsson S, Tesar V,
69. Kiykim AA, Horoz M, Gok E: Successful treatment of resistant Segelmark M, Pusey CD: Patients double-seropositive for ANCA
antiglomerular basement membrane antibody positivity with and anti-GBM antibodies have varied renal survival, frequency
mycophenolic acid. Intern Med 49: 577–580, 2010 of relapse, and outcomes compared to single-seropositive
70. Lazor R, Bigay-Gamé L, Cottin V, Cadranel J, Decaux O, Fellrath patients. Kidney Int 2017, in press
WnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 03/27/2023

JM, Cordier JF; Groupe d’Etudes et de Recherche sur les Maladies 89. Basford AW, Lewis J, Dwyer JP, Fogo AB: Membranous nephrop-
Orphelines Pulmonaires (GERMOP); Swiss Group for Interstitial athy with crescents. J Am Soc Nephrol 22: 1804–1808, 2011
and Orphan Lung Diseases (SIOLD): Alveolar hemorrhage in 90. Jia XY, Hu SY, Chen JL, Qu Z, Liu G, Cui Z, Zhao MH: The
anti-basement membrane antibody disease: A series of 28 cases. clinical and immunological features of patients with
Medicine (Baltimore) 86: 181–193, 2007 combined anti-glomerular basement membrane disease and
71. Herbert DG, Buscher H, Nair P: Prolonged venovenous extra- membranous nephropathy. Kidney Int 85: 945–952, 2014
corporeal membrane oxygenation without anticoagulation: A 91. Bandak G, Jones BA, Li J, Yee J, Umanath K: Rituximab for the
case of Goodpasture syndrome-related pulmonary haemor- treatment of refractory simultaneous anti-glomerular basement
rhage. Crit Care Resusc 16: 69–72, 2014 membrane (anti-GBM) and membranous nephropathy. Clin
72. Balke L, Both M, Arlt A, Rosenberg M, Bewig B: Severe adult Kidney J 7: 53–56, 2014
respiratory distress syndrome from Goodpasture syndrome. 92. Cui Z, Zhao MH, Singh AK, Wang HY: Antiglomerular basement
Survival using extracorporeal membrane oxygenation. Am J membrane disease with normal renal function. Kidney Int 72:
Respir Crit Care Med 191: 228–229, 2015 1403–1408, 2007
73. Legras A, Mordant P, Brechot N, Bel A, Boussaud V, Guillemain 93. McAdoo SP, Tanna A, Randone O, Tam FW, Tarzi RM, Levy JB,
R, Cholley B, Gibault L, Le Pimpec-Barthes F, Combes A: Pro- Griffith M, Lightstone L, Cook HT, Cairns T, Pusey CD: Necro-
longed extracorporeal membrane oxygenation and lung trans- tizing and crescentic glomerulonephritis presenting with pre-
plantation for isolated pulmonary anti-GBM (Goodpasture) served renal function in patients with underlying multisystem
disease. Intensive Care Med 41: 1866–1868, 2015 autoimmune disease: A retrospective case series. Rheumatology
74. Alchi B, Griffiths M, Sivalingam M, Jayne D, Farrington K: Pre- (Oxford) 54: 1025–1032, 2015
dictors of renal and patient outcomes in anti-GBM disease: 94. Troxell ML, Houghton DC: Atypical anti-glomerular basement
Clinicopathologic analysis of a two-centre cohort. Nephrol Dial membrane disease. Clin Kidney J 9: 211–221, 2016
Transplant 30: 814–821, 2015 95. Nasr SH, Collins AB, Alexander MP, Schraith DF, Herrera
75. Laczika K, Knapp S, Derfler K, Soleiman A, Hörl WH, Druml W: Hernandez L, Fidler ME, Sethi S, Leung N, Fervenza FC, Cornell
Immunoadsorption in Goodpasture’s syndrome. Am J Kidney LD: The clinicopathologic characteristics and outcome of
Dis 36: 392–395, 2000 atypical anti-glomerular basement membrane nephritis. Kidney
76. Liu P, Waheed S, Boujelbane L, Maursetter LJ: Multiple recur- Int 89: 897–908, 2016
rences of anti-glomerular basement membrane disease with 96. Reynolds J, Abbott DS, Karegli J, Evans DJ, Pusey CD: Mucosal
variable antibody detection: Can the laboratory be trusted? Clin tolerance induced by an immunodominant peptide from rat
Kidney J 9: 657–660, 2016 alpha3(IV)NC1 in established experimental autoimmune glo-
77. Gu B, Magil AB, Barbour SJ: Frequently relapsing anti- merulonephritis. Am J Pathol 174: 2202–2210, 2009
glomerular basement membrane antibody disease with 97. Reynolds J, Preston GA, Pressler BM, Hewins P, Brown M, Roth
changing clinical phenotype and antibody characteristics A, Alderman E, Bunch D, Jennette JC, Cook HT, Falk RJ, Pusey
over time. Clin Kidney J 9: 661–664, 2016 CD: Autoimmunity to the alpha 3 chain of type IV collagen in
78. Briggs JD, Jones E; Scientific Advisory Board of the ERA-EDTA glomerulonephritis is triggered by ‘autoantigen complemen-
Registry. European Renal Association-European Dialysis and tarity’. J Autoimmun 59: 8–18, 2015
Transplant Association: Renal transplantation for uncommon 98. McAdoo SP, Reynolds J, Bhangal G, Smith J, McDaid JP, Tanna A,
diseases. Nephrol Dial Transplant 14: 570–575, 1999 Jackson WD, Masuda ES, Cook HT, Pusey CD, Tam FW: Spleen
79. Sauter M, Schmid H, Anders HJ, Heller F, Weiss M, Sitter T: Loss tyrosine kinase inhibition attenuates autoantibody production
of a renal graft due to recurrence of anti-GBM disease despite and reverses experimental autoimmune GN. J Am Soc Nephrol
rituximab therapy. Clin Transplant 23: 132–136, 2009 25: 2291–2302, 2014
80. Byrne MC, Budisavljevic MN, Fan Z, Self SE, Ploth DW: Renal 99. Smith J, McDaid JP, Bhangal G, Chawanasuntorapoj R, Masuda
transplant in patients with Alport’s syndrome. Am J Kidney Dis ES, Cook HT, Pusey CD, Tam FWK: A spleen tyrosine kinase
39: 769–775, 2002 inhibitor reduces the severity of established glomerulonephritis.
81. Kashtan CE: Renal transplantation in patients with Alport syn- J Am Soc Nephrol 21: 231–236, 2010
drome. Pediatr Transplant 10: 651–657, 2006 100. McAdoo SP, Bhangal G, Page T, Cook HT, Pusey CD, Tam FW:
82. Browne G, Brown PA, Tomson CR, Fleming S, Allen A, Herriot R, Correlation of disease activity in proliferative glomerulone-
Pusey CD, Rees AJ, Turner AN: Retransplantation in Alport post- phritis with glomerular spleen tyrosine kinase expression. Kid-
transplant anti-GBM disease. Kidney Int 65: 675–681, 2004 ney Int 88: 52–60, 2015
83. Savige J, Gregory M, Gross O, Kashtan C, Ding J, Flinter F: Expert 101. Yang R, Otten MA, Hellmark T, Collin M, Björck L, Zhao MH,
guidelines for the management of Alport syndrome and thin Daha MR, Segelmark M: Successful treatment of experimental
basement membrane nephropathy. J Am Soc Nephrol 24: 364– glomerulonephritis with IdeS and EndoS, IgG-degrading strep-
375, 2013 tococcal enzymes. Nephrol Dial Transplant 25: 2479–2486,
84. Jayne DR, Marshall PD, Jones SJ, Lockwood CM: Autoantibodies 2010
to GBM and neutrophil cytoplasm in rapidly progressive glo- 102. McAdoo SP, Pusey CD: Anti-glomerular basement membrane
merulonephritis. Kidney Int 37: 965–970, 1990 disease. In: Encyclopedia of Medical Immunology: Autoim-
85. Levy JB, Hammad T, Coulthart A, Dougan T, Pusey CD: Clinical mune Diseases, edited by Mackay IR, Rose NR, Diamond B,
features and outcome of patients with both ANCA and anti-GBM Davidson A, New York, NY, Springer New York, 2014, pp 50–56
antibodies. Kidney Int 66: 1535–1540, 2004
86. Rutgers A, Slot M, van Paassen P, van Breda Vriesman P, Heeringa Published online ahead of print. Publication date available at www.
P, Tervaert JW: Coexistence of anti-glomerular basement cjasn.org.

You might also like