You are on page 1of 11

REVIEwS

Pathogenesis and therapeutic


interventions for ANCA-​associated
vasculitis
Daigo Nakazawa1, Sakiko Masuda2, Utano Tomaru3 and Akihiro Ishizu   2*
Abstract | Anti-​neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) affects
systemic small vessels and is accompanied by the presence of ANCAs in the serum. This disease
entity includes microscopic polyangiitis, granulomatosis with polyangiitis, eosinophilic
granulomatosis with polyangiitis and drug-​induced AAV. Similar to other autoimmune diseases,
AAV develops in patients with a predisposing genetic background who have been exposed to
causative environmental factors. The mechanism by which ANCAs cause vasculitis involves
ANCA-​mediated excessive activation of neutrophils that subsequently release inflammatory
cytokines, reactive oxygen species and lytic enzymes. In addition, this excessive activation of
neutrophils by ANCAs induces formation of neutrophil extracellular traps (NETs). Although NETs
are essential elements in innate immunity , excessive NET formation is harmful to small vessels.
Moreover, NETs are involved not only in ANCA-​mediated vascular injury but also in the
production of ANCAs themselves. Therefore, a vicious cycle of NET formation and ANCA
production is considered to be involved in the pathogenesis of AAV. In addition to this role of NETs
in AAV, some other important discoveries have been made in the past few years. Incorporating
these new insights into our understanding of the pathogenesis of AAV is needed to fully
understand and ultimately overcome this disease.

Following the initial discovery of anti-​neutrophil cytoplas- a granulomatous reaction that predominantly affects
mic antibodies (ANCAs) in 1982 (ref.1), ANCA-​associated small-​to-medium-​sized vessels2. This disease is associ-
vasculitis (AAV) was established as a disease entity dis- ated with adult-​onset asthma and allergic sinusitis; high
tinct from other vasculitides. AAV is now recognized as a numbers of eosinophils in peripheral blood and affected
systemic vasculitis of small vessels that is accompanied by tissues are its essential feature. About 50% of patients
the presence of ANCAs in the serum. The major antigens with EGPA are positive for MPO-​ANCAs2. Finally, drugs
targeted by these ANCAs are myeloperoxidase (MPO) such as propylthiouracil (an anti-​thyroid drug), hydral­
1
Department of
and proteinase 3 (PR3; also known as myeloblastin). azine (an anti-​hypertension drug) and cocaine can induce
Rheumatology, Endocrinology AAV includes microscopic polyangiitis (MPA), gran- production of ANCAs and lead to the development
and Nephrology, Faculty of ulomatosis with polyangiitis (GPA), eosinophilic granulo­ of drug-​induced AAV2. Epidemiological studies have
Medicine and Graduate matosis with polyangiitis (EGPA) and drug-​induced revealed ethnic differences in both ANCA specificity and
School of Medicine, Hokkaido
AAV2. MPA affects systemic small vessels — preferen- manifestations of AAV3–6. For example, PR3-ANCAs
University, Sapporo, Japan.
tially those of the renal glomeruli — and the majority and GPA are most common in Western countries,
2
Department of Medical
Laboratory Science, Faculty
of patients with MPA are positive for MPO-​ANCAs2. whereas MPO-​ANCAs and MPA predominate in East
of Health Sciences, Hokkaido The characteristic histopathological feature of MPA Asian countries (including Japan)3–6.
University, Sapporo, Japan. is necrotizing vasculitis; granulomatous inflammation is International collaborative groups have published
3
Department of Pathology, generally absent2. By contrast, GPA is characterized by guidelines for diagnosis 7,8 and treatments aimed at
Faculty of Medicine and granulomatous inflammation with necrosis that usually remission induction9,10 and maintenance of remission
Graduate School of Medicine, involves the respiratory tract and the simultaneous devel- of AAV11. Although these efforts have resulted in a
Hokkaido University,
Sapporo, Japan.
opment of necrotizing small-​vessel vasculitis2. Typically, remarkable improvement in the prognosis of affected
patients with GPA are positive for PR3-ANCAs2. Pauci-​ patients, more work is needed to fully understand and
*e-​mail: aishizu@
med.hokudai.ac.jp immune necrotizing and crescentic glomerulonephritis ultimately overcome AAV. Therefore, in this Review, we
https://doi.org/10.1038/ (NCGN) is associated with both MPA and GPA 2. present an updated understanding of the aetiology and
s41584-018-0145-y EGPA is an eosinophil-​rich necrotizing vasculitis with pathogenesis of AAV.

Nature Reviews | Rheumatology


Reviews

Epigenetic factors
Key points
Epigenetic modifications, including trimethylation of
• Anti-​neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) affects histone 3 lysine 27 (H3K27me3) and DNA methylation,
systemic small vessels and is accompanied by the presence of ANCAs in the serum. are implicated in the regulation of MPO and PRTN3
• AAV includes microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA), gene expression18,19. Reduced H3K27me3 is associated
eosinophilic granulomatosis with polyangiitis (EGPA) and drug-​induced AAV. with aberrant expression of MPO and PRTN3 genes
• AAV can develop in patients with a genetically predisposing background who are in patients with active AAV18. The levels of MPO and
exposed to causative environmental factors, such as infectious agents, drugs and air PRTN3 promoter methylation are negatively correlated
pollutants. with the number of mRNA transcripts of these genes19.
• ANCAs have a central role in the pathogenesis of AAV because they induce excessive Moreover, MPO and PRTN3 promoter methylation is
activation of neutrophils, which results in injury to small vessels. reduced in patients with active AAV and is increased
• Other immune cells (such as dendritic cells, macrophages, B cells and T cells), the during remission of AAV19. Elevated expression of MPO
complement system and humoral factors are also involved in the pathogenesis of AAV. and PR3, mediated by these epigenetic modifications, is
• Elucidation of the aetiology and pathogenesis of AAV is needed to develop new also linked with AAV19.
biomarkers as well as novel targeted therapeutic agents.
Environmental factors
Aetiology Environmental factors that can trigger the development
Genetic factors of AAV include infectious agents, drugs (such as pro-
Genome-​wide association studies (GWASs)12–14 have pylthiouracil, hydralazine and cocaine), and airborne
identified several genes involved in either susceptibility particulates such as silica dust. For example, toxic shock
or resistance to AAV (Table 1). Among these, the strongest syndrome toxin 1, which is secreted by Staphylococcus
associations with AAV are displayed by major histocom- aureus, is a risk factor for disease relapse in GPA20.
patibility complex class II (MHC II) genes. A GWAS of a Although the association of silica with AAV has been
European population demonstrated that GPA with PR3- intensely debated, a systematic review and meta-​analysis
ANCAs is most strongly associated with the HLA-DP confirmed the association between silica dust exposure
region and that MPA with MPO-​ANCAs is highly asso- and development of AAV21. Moreover, morbidity and
ciated with the HLA-​DQ region12. Interestingly, these disease severity increased substantially in patients with
data also indicated that the HLA genomic signature was AAV after the great earthquakes in Japan (Hanshin-​Awaji
associated with ANCA specificity (that is, PR3-ANCAs in 1995 (ref.22) and East Japan in 2011 (ref.23)). These
or MPO-​ANCAs) rather than with the clinical manifes- observations suggest that silica dust in the air, caused
tation (GPA or MPA). A GWAS of a North American by the massive destruction and subsequent reconstruc-
population also found that the HLA-​DP locus had the tion of the cities22 and by the marine sludge and sedi-
strongest association with GPA13. That study further ments deposited in the subsequent tsunami disaster23,
identified HLA-​DPB1*04 as a risk allele for GPA. By con- might have affected the presentation of AAV, espe-
trast, HLA-​DRB1*09:01, which is common in East Asian cially its respiratory manifestations such as pulmonary
but rare in European populations, is strongly associated haemorrhage and interstitial pneumonitis. However,
with MPA (MPO-​ANCAs) in the Japanese population15. in a contrasting study, no difference in the incidence
These ethnic differences in MHC II allele frequencies of AAV was observed before versus after the February
reflect the epidemiological differences observed in AAV; 2011 earthquake in Christchurch, New Zealand 24.
that is, the predominance of GPA with PR3-ANCAs in Therefore, whether environmental pollution from earth-
white European populations and the predominance of quakes can affect the incidence or presentation of AAV
MPA with MPO-​ANCAs in Asian populations3. remains unresolved.
Non-​MHC genes identified as associated with AAV
include PTPN22 (encoding tyrosine-​protein phos- Anti-​neutrophil cytoplasmic antibodies
phatase non-​receptor type 22)16, SERPINA1 (encoding The two major types of ANCAs have different cellular
α1-antitrypsin), PRTN3 (encoding PR3) and SEMA6A localization patterns, which can be detected by indirect
(encoding semaphorin 6a)12–14. The frequency of a gain-​ immunofluorescence of ethanol-​fixed neutrophils. One
of-function single-​nucleotide polymorphism (SNP) in type of ANCA is associated with staining around the
PTPN22 is higher in white patients with AAV than in the nucleus and is known as perinuclear ANCA (p-​ANCA),
general American white population 16. This variant whereas the other type is associated with diffuse staining
increases the activity of PTPN22, which negatively reg- of the cytoplasm and is known as cytoplasmic ANCA
ulates the production of IL-10 (an immuno­suppressive (c-​ANCA). The major antigen targeted by p-​ANCA is
cytokine), resulting in hyper-​responsiveness of the MPO and that targeted by c-​ANCA is PR3. At present,
immune system in patients with AAV16. A SNP near ANCAs are usually detected quantitatively by enzyme-​
SERPINA1 is associated with resistance to GPA (PR3- linked immunosorbent assay (ELISA) using plates
ANCA)17. Because α1-antitrypsin is an inhibitor of PR3, coated with these antigens. ANCAs detected by ELISA
this SNP might affect the function of PR3.17 A SNP in are called MPO-​ANCAs or PR3-ANCAs according to
PRTN3 is associated with resistance to PR3-ANCA the target antigen used.
AAV12, whereas a SNP in SEMA6A is associated with In addition to MPO and PR3, several other
resistance to GPA 13,14. Future studies are expected neutrophil-​d erived molecules can be targeted by
to reveal how these molecules are involved in the ANCAs, including α-​enolase, azurocidin, bactericidal
pathogenesis of AAV. permeability-​increasing protein (BPI), cathepsin G,

www.nature.com/nrrheum
Reviews

Table 1 | genes associated with aaV of NETs, which seems to be specific to some autoimmune
diseases. Although NETs are appropriately degraded by
gene associated oR Refs serum DNase I under physiological conditions36, NET
disease
degradation is reduced in patients with AAV42 as well
HL A-​DP • GPA • 5.39 12,13
as in patients with systemic lupus erythematosus 36.
• PR3-AAV • 7.03
The third step is antigen presentation. Susceptibility to
HL A-​DQ • MPA • 0.67 12
AAV is strongly associated with specific MHC II geno-
• MPO-​AAV • 0.65 types12,13,15. Therefore, patients who express MHC II mol-
HL A-​DR • MPA • 1.56 15
ecules that are apt to present MPO or PR3 are those most
• MPO-​AAV • 1.57 likely to produce ANCAs. This phenomenon could be
PTPN22 PR3-AAV 1.63 16
specific to patients with AAV. Misfolded MPO presented
SERPINA1 • GPA • 0.54 12 by MPA-​susceptible class II MHC (HLA-​DR) can also
• PR3-AAV • 0.53 be a target of ANCAs43. These misfolded MPO proteins
PRTN3 • GPA • 0.78 12 are transported to the cell surface (via a chaperone-​like
• PR3-AAV • 0.73 function of HLA-​DR), where cryptic epitopes of MPO
are recognized by the immune system43.
SEMA6A GPA 0.74 13,14

AAV, anti-​neutrophil-cytoplasmic-​antibody-associated vasculitis;


GPA , granulomatosis with polyangiitis; MPA , microscopic Pathogenicity of anti-​neutrophil cytoplasmic antibodies.
polyangiitis; MPO, myeloperoxidase; PR3, proteinase 3. Several animal models of AAV have been established,
including passive immune models44–48, active immune
elastase, defensin, lactoferrin, lysosome-​associated models49–53, drug-​induced models37,54, molecular mim-
membrane glycoprotein 2 (LAMP2) and moesin25–32. icry models27 and spontaneous models55. The character-
The pathogenicity of these ‘minor’ ANCAs is generally istics of these models are summarized in Supplementary
low, and p-​ANCAs other than MPO-​ANCAs are not Tables 1–3. Studies in these models have shown that
usually associated with vasculitis; however, discussion is ANCAs not only serve as a biomarker of AAV but
ongoing as to whether some minor ANCAs are involved also have pathogenic potential themselves. For exam-
in the pathogenesis of AAV25–32. ple, injection of either MPO-​ANCAs (obtained by the
immunization of MPO-​deficient mice with mouse
Mechanisms of anti-​neutrophil cytoplasmic antibody pro- MPO) or splenocytes from such immunized mice into
duction. Disordered regulation of neutrophil extracellular immunocompromised or wild-​type mice can induce
traps (NETs) is known to contribute to ANCA production. NCGN44. In other animal models, such as Wistar-​Kyoto
Although NETs are an element of the innate immune sys- rats immunized with human MPO, the anti-​human
tem that is essential for host defence33, the formation and MPO antibodies generated cross-​react with rat MPO,
breakdown of NETs is strictly regulated because exces- resulting in the development of NCGN and pulmonary
sive exposure to NETs can cause angiopathy34,35. NETs haemorrhage49. These animal models clearly indicate the
are degraded mainly by DNase I in serum36; however, the pathogenicity of ANCAs.
administration of propylthiouracil produces abnormal The mechanism by which ANCAs cause vasculitis has
NETs that are resistant to degradation by DNase I and thus been explained as follows56. Pro-​inflammatory cytokines
can persist in vivo37. Tolerance to MPO present in these such as TNF and IL-1β are generated during infection and
NETs is thereafter broken, resulting in MPO-​ANCA pro- activate neutrophils, which then express target antigens
duction. Of interest, MPO-​ANCAs are produced in ~30% (namely, MPO and PR3) on their cell surface. ANCAs
of patients treated with propylthiouracil38. The molecular bind to these antigens. At the same time, the crystalliza-
modification of MPO induced by propylthiouracil — a ble fragment (Fc) portion of these ANCAs binds to Fcγ
change in the protein structure surrounding the haem receptors on neutrophils, inducing excessive activation
iron from a rhombic to an axial form39 — could contribute of the neutrophils. This hyperactivation results in abnor-
to the breakdown of tolerance to MPO37. mal cytokine production accompanied by the release of
In mice, the injection of a mixture of myeloid den- reactive oxygen species (ROS) and lytic enzymes, which
dritic cells and neutrophils that have formed NETs injure vascular endothelial cells57,58. The excessive acti-
induces ANCA production, whereas the injection of a vation of neutrophils elicited by ANCAs also induces the
mixture of myeloid dendritic cells and DNase-​I-treated formation of NETs42,59. Moreover, humoral factors other
NET-​forming neutrophils does not induce ANCA pro- than ANCAs, such as damage-​associated molecular pat-
duction40. This finding suggests that the DNA in NETs terns, are involved in the formation of NETs in patients
is required for MPO-​ANCA and PR3-ANCA formation. with AAV60. This excessive NET formation — specifi-
The DNA in NETs can also activate B cells via Toll-​like cally, exposure to NET components such as histones
receptor 9 to accelerate antibody production41. and matrix metalloproteinases (MMPs) — is harmful to
In summary, several steps are involved in the process vascular endothelial cells34,35,56.
of ANCA production. During NET formation, the con- Despite the critical role of ANCAs in the pathogene-
tents of neutrophilic granules, including MPO and PR3, sis of AAV, the histological hallmark of AAV is necrotiz-
are mixed with chromatin fibres and bind to DNA, which ing glomerulonephritis without apparent deposition of
might modify the antigenicity of these autoantigens40. immune complexes (pauci-​immune glomerulonephritis).
This physiological first step is not specific to patients Of interest, when TNF-​primed neutrophils were exposed
with AAV. The second step is the incomplete degradation to ANCAs in  vitro, the ANCAs disappeared after

Nature Reviews | Rheumatology


Reviews

induction of NETs owing to their digestion by neutro- and low-​affinity antibodies69. Vasculitic disease activity
phil elastase derived from NET-​forming neutrophils61. is higher in patients with high-​affinity MPO-​ANCAs
These findings suggest that NETs contribute to the than in those with low-​affinity MPO-​ANCAs42,69. In
disappearance of immunoglobulins from AAV lesions. patients with low vasculitic disease activity despite
Immunofluorescence studies can detect NETs in the high MPO-ANCA titres, the MPO-​ANCAs might tar-
necrotizing lesions of AAV by the presence of citrulli- get epitopes located in a low-​risk region, instead of at
nated histones accompanied by extracellular DNA59,62–64. the amino terminus of the MPO heavy chain, or the
In addition, in our own unpublished research, we have MPO-ANCAs might have low antigen affinity.
found scattering of citrullinated histones, in a manner ANCAs cannot be detected in some patients with
that cannot be explained by an intracellular distribu- clinical manifestations of MPO-​AAV. In some of these
tion, in vessels affected by AAV (Fig. 1). Interestingly, patients, laboratory tests for ANCAs can give false-​
almost no NET deposition can be detected in the negative results because 50 kDa fragments of cerulo­
lesions of ANCA-​unrelated necrotizing vasculitis, such plasmin bind to the ANCAs in the serum70. However,
as polyarteritis nodosa (PAN). the clinical relevance of the production of ceruloplasmin
degradation products in vivo remains unclear.
Association of anti-​neutrophil cytoplasmic antibod-
ies with disease activity. Although AAV is an ANCA-​ Pathogenesis
associated disease, ANCA titres do not necessarily The pathogenesis of AAV partially overlaps between
reflect disease activity except for renal involvement65. MPA, GPA, EGPA and drug-​induced AAV. Of these,
The lack of correlation between ANCA titres and vas- MPA is considered the prototypic AAV because the
culitic disease activity can be explained by differences in pathways involved in its pathogenesis (Fig. 2) are actually
the epitopes and affinity of ANCAs. MPO is a 158 kDa shared by all types of AAV. Accordingly, the discussion of
heterotetrameric glycoprotein consisting of two light common pathways below, although particularly relevant
chains (14 kDa each), two heavy chains (59 kDa each) to MPA, also applies to the other subtypes of AAV.
and two haem molecules66. ANCAs in patients with
MPO-​AAV typically recognize an epitope at the amino Common pathways
terminus of the MPO heavy chain67,68; this specificity Priming of neutrophils. Upon the capture of infec-
is most likely in patients with the most severe disease tious agents, dendritic cells produce transforming
activity. MPO-​ANCAs are subdivided into high-​affinity growth factor-​β (TGFβ) and IL-6, which induce the

a b

Citrullinated histone 3 (NETs)


CD15 (neutrophils)
DAPI (DNA)

Fig. 1 | Nets are found in necrotizing lesions associated with mPa but not PaN. a | Formalin-​fixed, paraffin-​embedded
tissue sections of vasculitic lesions from (top) a patient with microscopic polyangiitis (MPA), the prototypic anti-​neutrophil
cytoplasmic antibody (ANCA)-associated vasculitis (AAV), and (bottom) a patient with polyarteritis nodosa (PAN), an
ANCA-​unrelated disease. b | Immunofluorescence images of vasculitic lesions from these patients reveal neutrophil
extracellular traps (NETs) in the patient with MPA (top) but not in the patient with PAN (bottom). Staining in the left panel is
with haematoxylin and eosin. In the right panel, the immunofluorescent stains used bind to citrullinated histone 3, a NET
marker ; and CD15, a neutrophil marker. 4ʹ,6-diamidino-2-phenylindole (DAPI) is an immunofluorescent stain that binds
strongly to adenine–thymine-​rich regions in DNA , commonly used as a DNA marker.

www.nature.com/nrrheum
Reviews

• TNF TH17 Naive T cell


• IL-1β
IL-17

Cytokine Dendritic
Macrophages • TGFβ cells
receptor
• IL-6
IL-23
C5a Infectious
Priming of agents
neutrophils
C5a C3bBb
receptor
C5-converting C3bB C3b C3
enzyme
C5 Factor D Factor B
Activation of complement Phagocytosis
alternative pathway by neutrophils
ANCA binding

• MPO
• PR3 ANCA

Fcγ
receptor

Plasma cells
BAFF IL-21

B cells

CD4+ T cells

Excessive activation NET


of neutrophils formation
MMPs
• ROS
• Lytic enzymes
Histone

Vascular endothelial cells


Development of vasculitis

Fig. 2 | Common pathways in the pathogenesis of aaV. Dendritic cells present antigens, such as those derived
from bacteria, to naive T cells, which differentiate into T helper 17 (TH17) cells and produce IL-17 A. Macrophages
stimulated by IL-17 A produce pro-​inflammatory cytokines, such as TNF and IL-1β, which prime neutrophils. At the
same time, C5a generated by activation of the alternative complement pathway binds to the C5a receptor on
the surface of neutrophils, which also primes neutrophils. Meanwhile, neutrophils stimulated by bacteria form
neutrophil extracellular traps (NETs). In patients with low NET degradation activity , these NETs persist, and
prolonged exposure to their contents disrupts tolerance to specific self-​antigens, notably myeloperoxidase (MPO)
and proteinase 3 (PR3). These antigens are presented to CD4+ T cells by dendritic cells, resulting in production of
anti-​neutrophil cytoplasmic antibodies (ANCAs). Primed neutrophils express MPO and PR3 on their plasma
membrane, to which PR3-ANCAs and MPO-​ANCAs bind; at the same time, the crystallizable fragment (Fc) region
of these ANCAs binds to the Fcγ receptor on neutrophils. This binding induces excessive activation of neutrophils,
leading to abnormal cytokine production accompanied by the release of reactive oxygen species (ROS) and lytic
enzymes, and further NET formation, which injures vascular endothelial cells. Humoral factors other than ANCAs,
such as damage-​associated molecular patterns (DAMPs), are also involved in NET formation in patients with
ANCA-​associated vasculitis (AAV). The major angiopathic molecules in NETs are histones dissociated from DNA
and matrix metalloproteinases (MMPs) such as MMP2 and MMP9. Both B cell-​activating factor (BAFF, also known
as B lymphocyte stimulator (BLyS) or TNF ligand superfamily member 13B), which is produced by activated
neutrophils, and CD4+ T cells (via IL-21) stimulate B cells. This process enables continuous ANCA production.
TGFβ, transforming growth factor-​β.

Nature Reviews | Rheumatology


Reviews

differentiation of naive T cells into T helper 17 (TH17) activated neutrophils is also involved in the activation
cells71. IL-17 (which is released from TH17 cells stim- of B cells82.
ulated by dendritic-​cell-derived IL-23) can induce the
release of pro-​inflammatory cytokines, such as TNF and Excessive activation of neutrophils. Primed neutrophils
IL-1β, from macrophages72. These pro-​inflammatory express ANCA-​specific antigens on their plasma mem-
cytokines prime neutrophils. brane. ANCAs bind to these antigens and, at the same
Disturbed T cell immune homeostasis is critically time, the Fc region of ANCAs binds to the Fcγ receptor
involved in both this priming and subsequent processes. on neutrophils. This binding induces excessive activa-
For example, functional and numerical abnormalities tion of these neutrophils, leading to abnormal cytokine
of regulatory T (Treg) cells, which suppress the prolif- production, the release of ROS and lytic enzymes, and
eration of autoreactive T cells, are associated with the eventually NET formation57–59,83. The most important
development of AAV73. In patients with AAV, a propor- angiopathic molecules in NETs are histones dissoci-
tion of Treg cells differentiate into TH17 cells, resulting in ated from DNA and MMPs such as MMP2 and MMP9
increased production of IL-17A (ref.72). In addition, low (refs34,35,56). Together, these data indicate that a vicious
expression of CD122 (IL-2 receptor β-​chain, an impor- cycle of NET and ANCA formation is involved in the
tant signal transducer of T cells) on CD4+CD25+ T cells pathogenesis of AAV (Fig. 3).
and Treg cells is associated with systemic vasculitis with Semaphorin 4D regulates neutrophil activation in
renal involvement and with relapse of AAV74. small vessels, and impairment of this regulation has
The complement system is also involved in priming of been demonstrated to lead to NET-​mediated vascular
neutrophils; accordingly, the role of complement proteins injury84,85. Serum levels of soluble semaphorin 4D are
in the pathogenesis of MPO-​AAV has been studied in elevated in patients with AAV and correlate with disease
mouse models. C4-deficient mice injected with MPO-​ activity84,85. The same researchers also found that the
ANCAs also develop vasculitis, as do wild-​type mice cell-​surface expression of semaphorin 4D is decreased
injected with MPO-​ANCAs; however, mice deficient in in neutrophils from patients with AAV as a consequence
both C5 and factor B do not develop vasculitis after the of the proteolytic cleavage of membrane semaphorin
injection of MPO-​ANCAs75. This finding suggests that 4D84,85. Interestingly, membranous semaphorin 4D on
the alternative complement pathway is involved in the neutrophils binds to plexin B2 on vascular endothelial
pathogenesis of MPO-​AAV. Interestingly, injection of cells, and this interaction decreases NET formation84,85.
MPO-​ANCAs into C6-deficient mice also results in vas- Collectively, these findings suggest that a physiolog-
culitis; therefore, an alternative pathway mechanism that ical mechanism that inhibits NET formation in small
is not mediated by the membrane attack complex (MAC) vessels is impaired in patients with AAV and that this
is likely to contribute to the development of MPO-​AAV76. impairment is associated with increased NET formation.
Neutrophil priming by C5a — an activated C5 fragment Some researchers have found that serum levels of
— is one such mechanism that deserves further consid- NETs do not correlate with either AAV disease activity86
eration56. Binding of C5a to its receptor on the neutrophil or ANCA titres60,87. However, a gold-​standard method
cell surface induces neutrophil activation and ANCA-​ for quantifying NETs in vivo has not yet been estab-
mediated glomerulonephritis in mice77. In addition, the lished88; therefore, future studies are needed to define
C5a-​induced release of tissue factor from neutrophils the association between NETs and AAV.
can promote hypercoagulability in patients with AAV78.
Furthermore, elevated serum levels of C3a and C5a, Granulomatosis with polyangiitis
which suggest activation of the alternative complement Neutrophil priming and ANCA-​mediated excessive
pathway and neutrophil priming via the C5a receptor, activation of neutrophils also occur in GPA, similarly
have been demonstrated in patients with active AAV79,80. to MPA. However, necrotizing granulomas in the res-
piratory tract and PR3-ANCA production are distinct
MPO-​ANCA production. When stimulated by bacte- features of GPA.
ria neutrophils form NETs, which are then degraded
by various enzymes, the most important of which (in Necrotizing granuloma formation. The mechanism of
serum) is DNase I36. Serum DNase I activity is markedly necrotizing granuloma formation is thought to involve
lower in patients with MPA than in healthy individuals42. infection, possibly by Staphylococcus aureus, which
In patients with MPA who have low NET degradation activates tissue-​resident macrophages in the bronchial
activity, NETs persist in vivo and tolerance to MPO is epithelium via Toll-​like receptors89. These macrophages
disrupted, resulting in MPO-​ANCA production81. The release pro-​inflammatory cytokines, including TNF and
structural modification of MPO proteins39 contributes IL-1β, which promote recruitment of neutrophils and
to this mechanism because these modified proteins can monocytes from the blood into the developing lesion.
be recognized as neoantigens. Recruited neutrophils that encounter microorganisms
Dendritic cells are involved in the presentation release ROS and lytic enzymes and undergo lysis, result-
of MPO contained in NETs to CD4+ T cells40. These ing in the formation of the necrotic core of the lesion89.
CD4+ T cells induce the differentiation of B cells into The recruited monocytes differentiate into macrophages
plasma cells that produce MPO-​ANCAs via the pro- that secrete IL-23, resulting in the differentiation of
duction of IL-21 (ref.82). TNF ligand superfamily mem- T cells towards a TH17 phenotype. IL-17 released from
ber 13B (also known as B cell-​activating factor (BAFF) TH17 cells is critically implicated in formation of the
or B lymphocyte stimulator (BLyS)) released from granuloma that surrounds the necrotic region89.

www.nature.com/nrrheum
Reviews

• Infectious agents Low DNase I activity


• Drugs
• Other environmental
factors

Persistence of NETs Excessive NET formation

• DAMPs
• Other humoral
factors
ANCA generation

Fig. 3 | a vicious cycle of Net and aNCa formation is involved in the pathogenesis of aaV. The activity of DNase I in
serum is considerably lower in patients with microscopic polyangiitis (MPA ; the prototypic anti-​neutrophil cytoplasmic
antibody (ANCA)-associated vasculitis (AAV)) than it is in healthy individuals. In consequence, when these patients are
exposed to precipitating environmental factors, such as infectious agents or drugs, breakdown of neutrophil extracellular
traps (NETs) by DNase I is impaired and the persistence of NET contents in vivo results in ANCA production. In turn, ANCAs
and other humoral factors (such as damage-​associated molecular patterns (DAMPs)) induce further NET formation; thus,
a NET–ANCA vicious cycle is accomplished.

PR3 on the surface of apoptotic neutrophils interferes to the human PRTN3 gene, which encodes PR3; thus,
with the induction of anti-​inflammatory mechanisms this observation suggests that the complementary PR3
following phagocytosis of these cells by macrophages90. transcripts have an exogenous origin. In support of this
In mouse models, concomitant injection of PR3-ANCAs notion, chronic carriage of S. aureus in the nasal cavity
and PR3-expressing apoptotic neutrophils induced a increases the risk of relapse of GPA96.
TH17 response, revealing a GPA-​specific mechanism
of immune polarization91. Furthermore, levels of IL-17 EGPA
and IL-23 are increased during active disease in patients EGPA is characterized by allergic manifestations such
with GPA92. Effector memory T cells, the proliferation of as adult-​onset asthma and sinusitis. Eosinophilia and
which is dependent on IL-15, also contribute to granu- a prominent infiltration of eosinophils into vasculitic
loma formation in patients with GPA93. PR3-stimulated lesions are distinct features of EGPA. About 50% of
dendritic cells derived from patients with GPA induced a patients with EGPA are positive for MPO-​ANCAs, and
higher IFNγ response in PR3-specific CD4+ T cells than the presence of these autoantibodies shows a positive
did PR3-stimulated dendritic cells from healthy individ- correlation with renal involvement but an inverse cor-
uals94. This TH1-type response might favour granuloma relation with cardiac involvement97. Interestingly, the
formation in patients with GPA. prevalence of ANCAs in patients with EGPA decreases
over time98.
PR3-ANCA production. MPO-​A NCAs and PR3-
ANCAs seem to share a fundamental underlying mech- Eosinophilia and infiltration of eosinophils. EGPA is
anism of production. Indeed, both types of ANCA are regarded as a TH2-cytokine-​mediated disease because
produced when mice are injected with NETs and mye- elevated levels of TH2 cytokines (such as IL-4, IL-5 and
loid dendritic cells40. The major factor that affects ANCA IL-13) are associated with eosinophilia in patients with
specificity seems to be the MHC II genotype, although EGPA99. C-​C motif chemokine 26 (CCL26; also known
other factors are specific to the production of each as eotaxin 3) released from vascular endothelial cells
type of ANCA. For example, a complementary PR3- is implicated in the tissue infiltration of eosinophils100.
mediated mechanism has been specifically implicated The tissue-​infiltrating eosinophils secrete eosinophilic
in the production of PR3-ANCAs in GPA95. The initial granules, including eosinophilic neurotoxin, major basic
immune response in GPA has been identified as directed proteins and eosinophilic cationic proteins, resulting in
against complementary PR3 transcripts, after which tissue destruction101. Although eosinophil peroxidase
PR3-ANCAs develop during a secondary anti-​idiotypic shares 68% amino acid identity with neutrophil MPO102,
immune response95. Interestingly, pathogens such as the mechanism of MPO-​ANCA production in patients
S. aureus have genetic sequences that are complementary with EGPA remains to be identified.

Nature Reviews | Rheumatology


Reviews

Anti-​lactoferrin antibody in EGPA. Anti-​lactoferrin prognosis of patients with AAV, development of thera-
antibodies (one of the ‘minor’ ANCAs) are present in a pies that target cells and molecules specifically involved
subgroup of patients with EGPA but not in patients with in the pathogenesis of AAV is needed to ultimately over-
MPA or GPA32. The frequency of renal involvement, come this disease. In the following sections, we focus on
serum C-​reactive protein levels and disease activity are the therapeutic possibilities inspired by discoveries in the
all considerably higher in anti-​lactoferrin-antibody-​ pathogenesis of AAV made in the past 5 years (Table 2).
positive than in anti-​lactoferrin-antibody-​negative
patients with EGPA. Lactoferrin is present in neutrophil-​ Targeting B cells
specific granules and is immediately secreted (via BAFF is produced by neutrophils in large amounts and has
degranulation) upon their activation. Lactoferrin is an important role in B cell survival. Serum levels of this
an endogenous suppressor of NET formation103; thus, molecule increase with progression of AAV108,109. A clin-
anti-​lactoferrin antibodies inhibit the NET-​suppressing ical trial of combination therapy with methotrexate plus
activity of lactoferrin and consequently promote NET the BAFF antagonist blisibimod had promising results110.
formation after activation of neutrophils32. Therefore, Bortezomib inhibits proteasomes, which have an
the presence of anti-​lactoferrin antibodies is thought essential role in the intracellular clearance of unwanted
to be associated with NET-​related disease activity in peptides. Treatment with bortezomib results in depletion
patients with EGPA. of antibody-​producing plasma cells. In a mouse model,
bortezomib treatment led to a reduction in MPO-​AAV
Drug-​induced AAV disease activity as a result of the deletion of MPO-​
Many drugs, such as propylthiouracil, benzylthiouracil, ANCA-producing plasma cells111. In humans, borte-
methimazole, carbimazole, minocycline, cefotaxime, zomib treatment ameliorates disease activity in patients
nitrofurantoin, d​-penicillamine, hydralazine, allopu- with refractory AAV112.
rinol, levamisole, phenytoin, sulfasalazine, rifampicin,
anti-​TNF agents and cocaine, can induce AAV104,105. Targeting T cells
Although several different pathways might lead to Cytotoxic lymphocyte antigen 4 (CTLA4), which is
the development of drug-​induced AAV, the innate expressed specifically and constitutively on Treg cells, also
and adaptive immune responses share some common has an important role in the regulation of self-​tolerance.
mechanisms. Abatacept, a selective co-​stimulation inhibitor, is a solu-
Propylthiouracil treatment induces the formation ble fusion protein composed of the Fc region of human
of DNase-​I-resistant NETs37. Because propylthiouracil IgG1 linked to the extracellular domain of CTLA4. This
does not inhibit DNase I activity directly, metabolites of agent binds to CD80 or CD86 on the surface of antigen-​
propylthiouracil are hypothesized to mask the DNase I presenting cells, thereby inhibiting T cell activation.
recognition sites in DNA extruded in NETs37. Cocaine A clinical study has demonstrated that abatacept treat-
and levamisole induce the formation of NETs enriched ment is well tolerated and associated with a high fre-
in neutrophil elastase and, potentially, in mitochondrial quency of disease remission in patients with relapsing
DNA, which is highly inflammatory106. Hydralazine also GPA113. The results of a double-​blind randomized con-
increases NET formation107, and NETs induced by these trolled trial of abatacept as first-​line therapy for AAV
drugs or reagents could be a source of antigens leading (ABAVAS)114 are expected to be released soon.
to ANCA formation. Some drugs, such as hydralazine,
induce promoter demethylation of DNA in T cells, result- Targeting cytokines
ing in their activation107. Activated T cells contribute to Patients with AAV can develop high levels of circulating
autoantibody production by B cells and plasma cells. cytokines. Serum levels of pro-​inflammatory cytokines,
particularly TNF and IL-6, are much higher in patients
Therapeutic targets in AAV with AAV than in healthy individuals115,116. Serum levels
International collaborative studies led to the publica- of IL-6 are also substantially increased in patients with
tion of treatment guidelines for both remission induc- active MPA and GPA115,116. Thus, treatment with anti-​
tion9,10 and maintenance of remission11. Although these cytokine biologic agents has been studied in patients
efforts have resulted in a remarkable improvement in the with these diseases.
Although conflicting reports have been published
on the efficacy of TNF blockade in patients with AAV
Table 2 | New therapeutic targets in aaV (reviewed elsewhere117), IL-6 blockade seems to be
target mechanism agent Refs a promising approach. IL-6 not only promotes B cell
B cells or plasma cells BAFF antagonist Blisibimod 110 differentiation but also has a central role in macrophage
activation, T cell differentiation, plasma cell survival and
Proteasome inhibitor Bortezomib 112
induction of other cytokines. Tocilizumab, a human-
T cells CD80 or CD86 antagonist Abatacept 113
ized anti-​IL-6 receptor antibody, achieved a complete
Cytokines Anti-​IL-6 receptor antibody Tocilizumab 117 and sustained remission in one patient with MPA
Anti-​IL-5 antibody Mepolizumab 118 who did not respond to standard immunotherapy118.
However, large-​s cale clinical studies are required
Complement system C5a receptor antagonist Avacopan 121
to define the adverse events associated with anti-​IL-6
Humoral factors Plasma exchange NA 122,123
therapy and to establish the long-​term prognosis of
NA , not applicable. treated patients.

www.nature.com/nrrheum
Reviews

IL-5 binds to the IL-5 receptor, which leads to the hospitalization124,125. A clinical trial of plasma exchange
growth of B cells and activation of eosinophils. A clini- and glucocorticoids (a cornerstone of AAV treatment126)
cal study has demonstrated that ~50% of patients with in patients with AAV (PEXIVAS)127 has been completed
EGPA treated with the anti-​IL-5 antibody mepolizumab and analysis of the results is now ongoing.
will enter remission119.
Conclusions
Targeting the complement system Thorough investigations into the genetic and envi-
The complement system is presumed to be crucial for the ronmental factors associated with AAV; ANCA path-
development of AAV. C5a, a product derived from acti- ogenicity; and the roles of neutrophils, other immune
vation of the alternative complement pathway, is thought cells and humoral factors in the pathogenesis of AAV
to be a major neutrophil-​activating molecule120. In mice, have led to several breakthroughs in the understanding
treatment with C5 and C5a receptor-​inhibiting antibod- and treatment of this disease. Consequently, promising
ies suppresses the development of MPO-​AAV76,121. The new therapeutic agents that target B cells, T cells and
C5a receptor antagonist avacopan blocks C5a-​mediated cytokines have been developed. However, no therapies
neutrophil activation and infiltration into the vascular have yet been developed that specifically target neutro-
endothelium. A clinical study of avacopan in patients phils, the pivotal contributors to this disease (although
with AAV demonstrated the therapeutic potency of glucocorticoids do prolong neutrophil survival)124.
this treatment, which was capable of replacing gluco- Future strategies should specifically address the role of
corticoids122. A larger clinical trial (ADVOCATE)123 is neutrophils in AAV. Finally, C5 and C5a receptor inhib-
underway that will Investigate the efficacy of this drug itors could become promising treatments for AAV, and
in patients with AAV. NET inhibitors (if they can be developed) might also be
potently effective. In the meantime, continued collab-
Targeting humoral factors orative studies as well as basic and clinical research are
Not only pathogenic ANCAs but also other humoral needed to fully understand the aetiology and pathogen-
factors are involved in the pathogenesis of AAV. Some esis of AAV and ultimately to develop safe and effective
clinical studies have demonstrated that plasma exchange, treatments.
which removes these factors, improves renal dysfunc-
tion in patients with AAV and shortens the duration of Published online xx xx xxxx

1. Davies, D. J., Moran, J. E., Niall, J. F. & Ryan, G. B. 13. Xie, G. et al. Association of granulomatosis with after the February 2011 Christchurch earthquake.
Segmental necrotising glomerulonephritis with polyangiitis (Wegener’s) with HLA-​DPB1*04 and Intern. Med. J. 47, 57–61 (2017).
antineutrophil antibody: possible arbovirus SEMA6A gene variants: evidence from genome-​wide 25. Moodie, F. D., Leaker, B., Cambridge, G., Totty, N. F. &
aetiology? Br. Med. J. (Clin. Res. Ed.) 285, 606 analysis. Arthritis Rheum. 65, 2457–2468 (2013). Segal, A. W. α-​enolase: a novel cytosolic autoantigen
(1982). 14. Rahmattulla, C. et al. Genetic variants in ANCA-​ in ANCA positive vasculitis. Kidney Int. 43, 675–681
2. Jennette, J. C. et al. 2012 revised International associated vasculitis: a meta-​analysis. Ann. Rheum. (1993).
Chapel Hill Consensus Conference nomenclature of Dis. 75, 1687–1692 (2016). 26. Yu, F. et al. Clinical and pathological features of renal
vasculitides. Arthritis Rheum. 65, 1–11 (2013). 15. Kawasaki, A. et al. Protective role of HLA-​ involvement in propylthiouracil-​associated ANCA-​
3. Fujimoto, S. et al. Comparison of the epidemiology of DRB1*13:02 against microscopic polyangiitis and positive vasculitis. Am. J. Kidney Dis. 49, 607–614
anti-​neutrophil cytoplasmic antibody-​associated MPO-​ANCA-positive vasculitides in a Japanese (2007).
vasculitis between Japan and the U.K. Rheumatology population: a case–control study. PLOS ONE 11, 27. Kain, R. et al. Molecular mimicry in pauci-​immune
50, 1916–1920 (2011). e0154393 (2016). focal necrotizing glomerulonephritis. Nat. Med. 14,
4. Furuta, S. et al. Comparison of phenotype and 16. Cao, Y. et al. High basal activity of the PTPN22 gain-​ 1088–1096 (2008).
outcome in microscopic polyangiitis between Europe of-function variant blunts leukocyte responsiveness 28. Nagao, T. et al. Direct activation of glomerular
and Japan. J. Rheumatol. 41, 325–333 (2014). negatively affecting IL-10 production in ANCA endothelial cells by anti-​moesin activity of anti-​
5. Furuta, S. et al. Comparison of the phenotype and vasculitis. PLOS ONE 7, e42783 (2012). myeloperoxidase antibody. Nephrol. Dial. Transplant.
outcome of granulomatosis with polyangiitis between 17. Relle, M., Fohr, B., Fasola, F. & Schwarting, A. Genetics 26, 2752–2760 (2011).
UK and Japanese cohorts. J. Rheumatol. 44, and pathophysiology of granulomatosis with polyangiitis 29. Kontic, M., Radovanovic, S., Nikolic, M.
216–222 (2017). (GPA) and its main autoantigen proteinase 3. Mol. Cell. & Bonaci-Nikolic, B. Concomitant drug- and infection-​
6. Pearce, F. A., Craven, A., Merkel, P. A., Probes 30, 366–373 (2016). induced antineutrophil cytoplasmic autoantibody
Luqmani, R. A. & Watts, R. A. Global ethnic and 18. Ciavatta, D. J. et al. Epigenetic basis for aberrant (ANCA)-associated vasculitis with multispecific ANCA.
geographic differences in the clinical presentations upregulation of autoantigen genes in humans with Med. Princ. Pract. 21, 488–491 (2012).
of anti-​neutrophil cytoplasm antibody-​associated ANCA vasculitis. J. Clin. Invest. 120, 3209–3219 30. Fukuhara, A. et al. Systemic vasculitis associated with
vasculitis. Rheumatology 56, 1962–1969 (2010). anti-​neutrophil cytoplasmic antibodies against
(2017). 19. Jones, B. E. et al. Gene-​specific DNA methylation bactericidal/permeability increasing protein.
7. Seeliger, B. et al. Are the 1990 American College changes predict remission in patients with ANCA-​ Intern. Med. 52, 1095–1099 (2013).
of Rheumatology vasculitis classification criteria associated vasculitis. J. Am. Soc. Nephrol. 28, 31. Suzuki, K. et al. A novel autoantibody against moesin
still valid? Rheumatology 56, 1154–1161 1175–1187 (2017). in the serum of patients with MPO-​ANCA-associated
(2017). 20. Popa, E. R. et al. Staphylococcal toxic-​shock- vasculitis. Nephrol. Dial. Transplant. 29, 1168–1177
8. Bossuyt, X. et al. Position paper: revised 2017 syndrome-​toxin-1 as a risk factor for disease relapse (2014).
international consensus on testing of ANCAs in in Wegener’s granulomatosis. Rheumatology 46, 32. Shida, H. et al. The presence of anti-​lactoferrin
granulomatosis with polyangiitis and microscopic 1029–1033 (2007). antibodies in a subgroup of eosinophilic
polyangiitis. Nat. Rev. Rheumatol. 13, 683–692 21. Gomez-​Puerta, J. A., Gedmintas, L. & Costenbader, K. H. granulomatosis with polyangiitis patients and their
(2017). The association between silica exposure and possible contribution to enhancement of neutrophil
9. Ntatsaki, E. et al. BSR and BHPR guideline for the development of ANCA-​associated vasculitis: extracellular trap formation. Front. Immunol. 7, 636
management of adults with ANCA-​associated systematic review and meta-​analysis. Autoimmun. Rev. (2016).
vasculitis. Rheumatology 53, 2306–2309 (2014). 12, 1129–1135 (2013). 33. Brinkmann, V. et al. Neutrophil extracellular traps kill
10. Yates, M. et al. EULAR/ERA-​EDTA recommendations 22. Yashiro, M. et al. Significantly high regional morbidity bacteria. Science 303, 1532–1535 (2004).
for the management of ANCA-​associated vasculitis. of MPO-​ANCA-related angitis and/or nephritis with 34. Grayson, P. C. & Kaplan, M. J. At the bench:
Ann. Rheum. Dis. 75, 1583–1594 (2016). respiratory tract involvement after the 1995 great neutrophil extracellular traps (NETs) highlight novel
11. Puechal, X. et al. Long-​term outcomes among earthquake in Kobe (Japan). Am. J. Kidney Dis. 35, aspects of innate immune system involvement in
participants in the WEGENT trial of remission-​ 889–895 (2000). autoimmune diseases. J. Leukoc. Biol. 99, 253–264
maintenance therapy for granulomatosis with 23. Takeuchi, Y. et al. The influence of the great East (2016).
polyangiitis (Wegener’s) or microscopic polyangiitis. Japan earthquake on microscopic polyangiitis: 35. Schreiber, A. et al. Necroptosis controls NET
Arthritis Rheumatol. 68, 690–701 (2016). a retrospective observational study. PLOS ONE 12, generation and mediates complement activation,
12. Lyons, P. A. et al. Genetically distinct subsets within e0177482 (2017). endothelial damage, and autoimmune vasculitis.
ANCA-​associated vasculitis. N. Engl. J. Med. 367, 24. Farquhar, H. J. et al. Incidence of anti-​neutrophil Proc. Natl Acad. Sci. USA 114, E9618–E9625
214–223 (2012). cytoplasmic antibody-​associated vasculitis before and (2017).

Nature Reviews | Rheumatology


Reviews

36. Hakkim, A. et al. Impairment of neutrophil 57. Falk, R. J., Terrell, R. S., Charles, L. A. & Jennette, J. C. 80. Gou, S. J., Yuan, J., Chen, M., Yu, F. & Zhao, M. H.
extracellular trap degradation is associated with lupus Anti-​neutrophil cytoplasmic autoantibodies induce Circulating complement activation in patients with
nephritis. Proc. Natl Acad. Sci. USA 107, 9813–9818 neutrophils to degranulate and produce oxygen anti-​neutrophil cytoplasmic antibody-​associated
(2010). radicals in vitro. Proc. Natl Acad. Sci. USA 87, vasculitis. Kidney Int. 83, 129–137 (2013).
37. Nakazawa, D. et al. Abnormal conformation and 4115–4119 (1990). 81. Nakazawa, D., Tomaru, U. & Ishizu, A. Possible
impaired degradation of propylthiouracil-​induced 58. Charles, L. A., Caldas, M. L., Falk, R. J., Terrell, R. S. & implication of disordered neutrophil extracellular
neutrophil extracellular traps: implications of Jennette, J. C. Antibodies against granule proteins traps in the pathogenesis of MPO-​ANCA-associated
disordered neutrophil extracellular traps in a rat activate neutrophils in vitro. J. Leukoc. Biol. 50, vasculitis. Clin. Exp. Nephrol. 17, 631–633 (2013).
model of myeloperoxidase antineutrophil cytoplasmic 539–546 (1991). 82. Kallenberg, C. G. Pathogenesis and treatment of
antibody-​associated vasculitis. Arthritis Rheum. 64, 59. Kessenbrock, K. et al. Netting neutrophils in ANCA-​associated vasculitides. Clin. Exp. Rheumatol.
3779–3787 (2012). autoimmune small-​vessel vasculitis. Nat. Med. 15, 33 (Suppl. 92), S11–S14 (2015).
38. Wada, N. et al. Prevalence of serum anti-​ 623–625 (2009). 83. Nakazawa, D. et al. The responses of macrophages
myeloperoxidase antineutrophil cytoplasmic 60. Kraaij, T. et al. Excessive neutrophil extracellular trap in  interaction with neutrophils that undergo NETosis.
antibodies (MPO-​ANCA) in patients with Graves’ formation in ANCA-​associated vasculitis is J. Autoimmun. 67, 19–28 (2016).
disease treated with propylthiouracil and thiamazole. independent of ANCA. Kidney Int. 94, 139–149 84. Nishide, M. et al. Semaphorin 4D inhibits neutrophil
Endocr. J. 49, 329–334 (2002). (2018). activation and is involved in the pathogenesis of
39. Lee, E. et al. Inactivation of peroxidases of rat bone 61. Futamata, E. et al. Vanishing immunoglobulins: the neutrophil-​mediated autoimmune vasculitis.
marrow by repeated administration of propylthiouracil formation of pauci-​immune lesions in Ann. Rheum. Dis. 76, 1440–1448 (2017).
is accompanied by a change in the heme structure. myeloperoxidase-​antineutrophil cytoplasmic antibody-​ 85. Nishide, M. & Kumanogoh, A. The role of semaphorins
Biochem. Pharmacol. 37, 2151–2153 (1988). associated vasculitis. Nephron 138, 328–330 (2018). in immune responses and autoimmune rheumatic
40. Sangaletti, S. et al. Neutrophil extracellular traps 62. Yoshida, M., Sasaki, M., Sugisaki, K., Yamaguchi, Y. diseases. Nat. Rev. Rheumatol. 14, 19–31 (2018).
mediate transfer of cytoplasmic neutrophil antigens to & Yamada, M. Neutrophil extracellular trap 86. Wang, H. et al. Circulating level of neutrophil
myeloid dendritic cells toward ANCA induction and components in fibrinoid necrosis of the kidney with extracellular traps is not a useful biomarker for
associated autoimmunity. Blood 120, 3007–3018 myeloperoxidase-​ANCA-associated vasculitis. assessing disease activity in antineutrophil
(2012). Clin. Kidney J. 6, 308–312 (2013). cytoplasmic antibody-​associated vasculitis. PLOS ONE
41. Hurtado, P. R. et al. CpG oligodeoxynucleotide 63. O’Sullivan, K. M. et al. Renal participation of 11, e0148197 (2016).
stimulates production of anti-​neutrophil cytoplasmic myeloperoxidase in antineutrophil cytoplasmic 87. Heeringa, P., Rutgers, A. & Kallenberg, C. G. M.
antibodies in ANCA associated vasculitis. BMC antibody (ANCA)-associated glomerulonephritis. The net effect of ANCA on neutrophil extracellular trap
Immunol. 9, 34 (2008). Kidney Int. 88, 1030–1046 (2015). formation. Kidney Int. 94, 14–16 (2018).
42. Nakazawa, D. et al. Enhanced formation and 64. Gupta, S. & Kaplan, M. J. The role of neutrophils and 88. Masuda, S. et al. NETosis markers: quest for specific,
disordered regulation of NETs in myeloperoxidase-​ NETosis in autoimmune and renal diseases. Nat. Rev. objective, and quantitative markers. Clin. Chim. Acta
ANCA-associated microscopic polyangiitis. J. Am. Soc. Nephrol. 12, 402–413 (2016). 459, 89–93 (2016).
Nephrol. 25, 990–997 (2014). 65. Fussner, L. A. et al. Factors determining the clinical 89. Kallenberg, C. G., Stegeman, C. A., Abdulahad, W. H.
43. Hiwa, R. et al. Myeloperoxidase/HLA class II utility of serial measurements of antineutrophil & Heeringa, P. Pathogenesis of ANCA-​associated
complexes recognized by autoantibodies in cytoplasmic antibodies targeting proteinase 3. vasculitis: new possibilities for intervention.
microscopic polyangiitis. Arthritis Rheumatol. 69, Arthritis Rheumatol. 68, 1700–1710 (2016). Am. J. Kidney Dis. 62, 1176–1187 (2013).
2069–2080 (2017). 66. Fiedler, T. J., Davey, C. A. & Fenna, R. E. X-​Ray crystal 90. Kantari, C. et al. Proteinase 3, the Wegener
44. Xiao, H. et al. Antineutrophil cytoplasmic structure and characterization of halide-​binding sites autoantigen, is externalized during neutrophil
autoantibodies specific for myeloperoxidase cause of human myeloperoxidase at 1.8|Å resolution. J. Biol. apoptosis: evidence for a functional association with
glomerulonephritis and vasculitis in mice. J. Clin. Chem. 275, 11964–11971 (2000). phospholipid scramblase 1 and interference with
Invest. 110, 955–963 (2002). 67. Suzuki, K. et al. Analysis of risk epitopes of anti-​ macrophage phagocytosis. Blood 110, 4086–4095
45. Huugen, D. et al. Aggravation of anti-​myeloperoxidase neutrophil antibody MPO-​ANCA in vasculitis in (2007).
antibody-​induced glomerulonephritis by bacterial Japanese population. Microbiol. Immunol. 51, 91. Millet, A. et al. Proteinase 3 on apoptotic cells
lipopolysaccharide: role of tumor necrosis factor-​α. 1215–1220 (2007). disrupts immune silencing in autoimmune vasculitis.
Am. J. Pathol. 167, 47–58 (2005). 68. Gou, S. J., Xu, P. C., Chen, M. & Zhao, M. H. Epitope J. Clin. Invest. 125, 4107–4121 (2015).
46. Kanzaki, G. et al. Impact of anti-​glomerular basement analysis of anti-​myeloperoxidase antibodies in 92. Nogueira, E. et al. Serum IL-17 and IL-23 levels and
membrane antibodies and glomerular neutrophil patients with ANCA-​associated vasculitis. PLOS ONE autoantigen-​specific TH17 cells are elevated in patients
activation on glomerulonephritis in experimental 8, e60530 (2013). with ANCA-​associated vasculitis. Nephrol. Dial.
myeloperoxidase-​antineutrophil cytoplasmic antibody 69. Yoshida, M. et al. Two types of myeloperoxidase-​ Transplant. 25, 2209–2217 (2010).
vasculitis. Nephrol. Dial. Transplant. 31, 574–585 antineutrophil cytoplasmic autoantibodies with a high 93. Lepse, N., Abdulahad, W. H., Kallenberg, C. G. &
(2016). affinity and a low affinity in small vessel vasculitis. Heeringa, P. Immune regulatory mechanisms in ANCA-​
47. Primo, V. C. et al. Anti-​PR3 immune responses Clin. Exp. Rheumatol. 27, S28–S32 (2009). associated vasculitides. Autoimmun. Rev. 11, 77–83
induce segmental and necrotizing glomerulonephritis. 70. Roth, A. J. et al. Epitope specificity determines (2011).
Clin. Exp. Immunol. 159, 327–337 (2010). pathogenicity and detectability in ANCA-​associated 94. Csernok, E. et al. Wegener autoantigen induces
48. Little, M. A. et al. Anti-​proteinase 3 anti-​neutrophil vasculitis. J. Clin. Invest. 123, 1773–1783 (2013). maturation of dendritic cells and licenses them for TH1
cytoplasm autoantibodies recapitulate systemic 71. Flint, S. M., McKinney, E. F. & Smith, K. G. Emerging priming via the protease-​activated receptor-2
vasculitis in mice with a humanized immune system. concepts in the pathogenesis of antineutrophil pathway. Blood 107, 4440–4448 (2006).
PLOS ONE 7, e28626 (2012). cytoplasmic antibody-​associated vasculitis. Curr. Opin. 95. Pendergraft, W. F. 3rd et al. Autoimmunity is triggered
49. Little, M. A. et al. Experimental autoimmune Rheumatol. 27, 197–203 (2015). by cPR-3105–201, a protein complementary to human
vasculitis: an animal model of anti-​neutrophil 72. Abdulahad, W. H., Lamprecht, P. & Kallenberg, C. G. autoantigen proteinase-3. Nat. Med. 10, 72–79 (2004).
cytoplasmic autoantibody-​associated systemic T-Helper cells as new players in ANCA-​associated 96. Stegeman, C. A. et al. Association of chronic nasal
vasculitis. Am. J. Pathol. 174, 1212–1220 (2009). vasculitides. Arthritis Res. Ther. 13, 236 (2011). carriage of Staphylococcus aureus and higher relapse
50. Brouwer, E. et al. Antimyeloperoxidase-​associated 73. Chavele, K. M. et al. Regulation of myeloperoxidase-​ rates in Wegener granulomatosis. Ann. Intern. Med.
proliferative glomerulonephritis: an animal model. specific T cell responses during disease remission in 120, 12–17 (1994).
J. Exp. Med. 177, 905–914 (1993). antineutrophil cytoplasmic antibody-​associated 97. Comarmond, C. et al. Eosinophilic granulomatosis with
51. Heeringa, P. et al. Autoantibodies to myeloperoxidase vasculitis: the role of Treg cells and tryptophan polyangiitis (Churg–Strauss): clinical characteristics
aggravate mild anti-​glomerular-basement-​membrane- degradation. Arthritis Rheum. 62, 1539–1548 and long-​term followup of the 383 patients enrolled in
mediated glomerular injury in the rat. Am. J. Pathol. (2010). the French Vasculitis Study Group cohort. Arthritis
149, 1695–1706 (1996). 74. Wilde, B., Hoerning, A., Kribben, A., Witzke, O. & Rheum. 65, 270–281 (2013).
52. Ruth, A. J. et al. Anti-​neutrophil cytoplasmic Dolff, S. Abnormal expression pattern of the IL-2 98. Bremer, J. P., Csernok, E., Holle, J., Gross, W. L. &
antibodies and effector CD4+ cells play nonredundant receptor β-​chain on CD4+ T cells in ANCA-​associated Moosig, F. Getting rid of MPO-​ANCA: a matter of
roles in anti-​myeloperoxidase crescentic vasculitis. Dis. Markers 2014, 249846 (2014). disease subtype. Rheumatology 52, 752–754 (2013).
glomerulonephritis. J. Am. Soc. Nephrol. 17, 75. Xiao, H., Schreiber, A., Heeringa, P., Falk, R. J. & 99. Khoury, P., Grayson, P. C. & Klion, A. D. Eosinophils in
1940–1949 (2006). Jennette, J. C. Alternative complement pathway in the vasculitis: characteristics and roles in pathogenesis.
53. Schreiber, A., Xiao, H., Falk, R. J. & Jennette, J. C. pathogenesis of disease mediated by anti-​neutrophil Nat. Rev. Rheumatol. 10, 474–483 (2014).
Bone marrow-​derived cells are sufficient and cytoplasmic autoantibodies. Am. J. Pathol. 170, 100. Polzer, K. et al. Eotaxin-3 is involved in Churg–Strauss
necessary targets to mediate glomerulonephritis and 52–64 (2007). syndrome — a serum marker closely correlating with
vasculitis induced by anti-​myeloperoxidase 76. Xiao, H. et al. C5a receptor (CD88) blockade protects disease activity. Rheumatology 47, 804–808 (2008).
antibodies. J. Am. Soc. Nephrol. 17, 3355–3364 against MPO-​ANCA GN. J. Am. Soc. Nephrol. 25, 101. Vaglio, A., Buzio, C. & Zwerina, J. Eosinophilic
(2006). 225–231 (2014). granulomatosis with polyangiitis (Churg–Strauss):
54. Kusunoki, Y. et al. Peptidylarginine deiminase inhibitor 77. Schreiber, A. et al. C5a receptor mediates neutrophil state of the art. Allergy 68, 261–273 (2013).
suppresses neutrophil extracellular trap formation and activation and ANCA-​induced glomerulonephritis. 102. Sullivan, S., Salapow, M. A., Breen, R. & Broide, D. H.
MPO-​ANCA production. Front. Immunol. 7, 227 J. Am. Soc. Nephrol. 20, 289–298 (2009). Eosinophil peroxidase differs from neutrophil
(2016). 78. Huang, Y. M., Wang, H., Wang, C., Chen, M. myeloperoxidase in its ability to bind antineutrophil
55. Kinjoh, K., Kyogoku, M. & Good, R. A. Genetic & Zhao, M. H. Promotion of hypercoagulability in cytoplasmic antibodies reactive with myeloperoxidase.
selection for crescent formation yields mouse strain antineutrophil cytoplasmic antibody-​associated Int. Arch. Allergy Immunol. 105, 150–154 (1994).
with rapidly progressive glomerulonephritis and small vasculitis by C5a-​induced tissue factor-​expressing 103. Okubo, K. et al. Lactoferrin suppresses neutrophil
vessel vasculitis. Proc. Natl Acad. Sci. USA 90, microparticles and neutrophil extracellular traps. extracellular traps release in inflammation.
3413–3417 (1993). Arthritis Rheumatol. 67, 2780–2790 (2015). EBioMedicine 10, 204–215 (2016).
56. Jennette, J. C. & Falk, R. J. Pathogenesis of 79. Yuan, J. et al. C5a and its receptors in human anti-​ 104. Grau, R. G. Drug-​induced vasculitis: new insights and a
antineutrophil cytoplasmic autoantibody-​mediated neutrophil cytoplasmic antibody (ANCA)-associated changing lineup of suspects. Curr. Rheumatol. Rep.
disease. Nat. Rev. Rheumatol. 10, 463–473 (2014). vasculitis. Arthritis Res. Ther. 14, R140 (2012). 17, 71 (2015).

www.nature.com/nrrheum
Reviews

105. Olives, T. D., Kornas, R. L., Fujisawa, R. & Cole, J. B. 114. US National Library of Medicine. ClinicalTrials.gov 125. Uechi, E., Okada, M. & Fushimi, K. Effect of plasma
Unexpected complication of cocaine-​associated https://clinicaltrials.gov/ct2/show/NCT00482066 exchange on in-​hospital mortality in patients with
anti-neutrophil cytoplasmic antibody vasculitis related (2015). pulmonary hemorrhage secondary to antineutrophil
to persistent in-​hospital cocaine use. J. Addict. Med. 115. Arimura, Y. et al. Serum myeloperoxidase and serum cytoplasmic antibody-​associated vasculitis: a propensity-​
11, 157–160 (2017). cytokines in anti-​myeloperoxidase antibody-​associated matched analysis using a nationwide administrative
106. Lood, C. & Hughes, G. C. Neutrophil extracellular glomerulonephritis. Clin. Nephrol. 40, 256–264 (1993). database. PLOS ONE 13, e0196009 (2018).
traps as a potential source of autoantigen in cocaine-​ 116. Ohlsson, S., Wieslander, J. & Segelmark, M. 126. Espinasse, M. A. et al. Glucocorticoid-​induced leucine
associated autoimmunity. Rheumatology 56, Circulating cytokine profile in anti-​neutrophilic zipper is expressed in human neutrophils and
638–643 (2017). cytoplasmatic autoantibody-​associated vasculitis: promotes apoptosis through Mcl-1 down-​regulation.
107. Irizarry-​Caro, J. A. et al. Brief report: drugs implicated prediction of outcome? Mediators Inflamm. 13, J. Innate Immun. 8, 81–96 (2016).
in systemic autoimmunity modulate neutrophil 275–283 (2004). 127. US National Library of Medicine. ClinicalTrials.gov
extracellular trap formation. Arthritis Rheumatol. 70, 117. McAdoo, S. P. & Pusey, C. D. Is there a role for TNFα https://clinicaltrials.gov/ct2/show/NCT00987389
468–474 (2018). blockade in ANCA-​associated vasculitis and (2018).
108. Schneeweis, C. et al. Increased levels of BLyS and glomerulonephritis? Nephrol. Dial. Transplant. 32,
sVCAM-1 in anti-​neutrophil cytoplasmatic antibody i80–i88 (2017). Acknowledgements
(ANCA)-associated vasculitides (AAV). Clin. Exp. 118. Berti, A. et al. Interleukin-6 in ANCA-​associated The authors’ research was supported by a grant from the
Rheumatol. 28, 62–66 (2010). vasculitis: rationale for successful treatment with Japan Research Committee of the Ministry of Health, Labour
109. Bader, L., Koldingsnes, W. & Nossent, J. B-Lymphocyte tocilizumab. Semin. Arthritis Rheum. 45, 48–54 and Welfare for intractable vasculitis.
activating factor levels are increased in patients with (2015).
Wegener’s granulomatosis and inversely correlated 119. Wechsler, M. E. et al. Mepolizumab or placebo for Author contributions
with ANCA titer. Clin. Rheumatol. 29, 1031–1035 eosinophilic granulomatosis with polyangiitis. N. Engl. A.I. wrote the manuscript and reviewed or edited the manu-
(2010). J. Med. 376, 1921–1932 (2017). script before submission. D.N., S.M. and U.T. contributed to
110. Lenert, A. & Lenert, P. Current and emerging 120. Chen, M., Jayne, D. R. W. & Zhao, M. H. Complement researching data for the article and discussions of the article
treatment options for ANCA-​associated vasculitis: in ANCA-​associated vasculitis: mechanisms and content.
potential role of belimumab and other BAFF/APRIL implications for management. Nat. Rev. Nephrol. 13,
targeting agents. Drug Des. Devel. Ther. 9, 333–347 359–367 (2017). Competing interests
(2015). 121. Huugen, D. et al. Inhibition of complement factor C5 The authors declare no competing interests.
111. Bontscho, J. et al. Myeloperoxidase-​specific plasma protects against anti-​myeloperoxidase antibody-​
cell depletion by bortezomib protects from anti-​ mediated glomerulonephritis in mice. Kidney Int. 71, Publisher’s note
neutrophil cytoplasmic autoantibodies-​induced 646–654 (2007). Springer Nature remains neutral with regard to jurisdictional
glomerulonephritis. J. Am. Soc. Nephrol. 22, 122. Jayne, D. R. W. et al. Randomized trial of C5a receptor claims in published maps and institutional affiliations.
336–348 (2011). inhibitor avacopan in ANCA-​associated vasculitis.
112. Novikov, P., Moiseev, S., Bulanov, N. & Shchegoleva, E. J. Am. Soc. Nephrol. 28, 2756–2767 (2017). Reviewer information
Bortezomib in refractoryANCA-​associated 123. US National Library of Medicine. ClinicalTrials.gov Nature Reviews Rheumatology thanks F. Moosig and
vasculitis: a new option? Ann. Rheum. Dis. 75, e9 https://clinicaltrials.gov/ct2/show/NCT02994927 A. Rutgers, and other anonymous reviewers, for their
(2016). (2018). contribution to the peer review of this work.
113. Langford, C. A. et al. An open-​label trial of abatacept 124. de Luna, G. et al. Plasma exchanges for the treatment
(CTLA4–Ig) in non-​severe relapsing granulomatosis of severe systemic necrotizing vasculitides in clinical Supplementary information
with polyangiitis (Wegener’s). Ann. Rheum. Dis. 73, daily practice: data from the French Vasculitis Study Supplementary information is available for this paper at
1376–1379 (2014). Group. J. Autoimmun. 65, 49–55 (2015). https://doi.org/10.1038/s41584-018-0145-y.

Nature Reviews | Rheumatology

You might also like