Clinical Practice: Review Article
Nephron 2022;146:121–137
Antineutrophil Cytoplasmic
Antibody-Associated Vasculitis: Toward
an Individualized Approach
Javier Villacorta 
a
Laura Martinez-Valenzuela 
b
Irene Martin-Capon 
a
Juliana Bordignon-Draibe 
b
aNephrology Department, Ramón y Cajal University Hospital, IRYCIS, Alcala University, Madrid, Spain;
b
Nephrology Department, Bellvitge University Hospital, IDIBELL Biomedical Research Institute, Hospitalet de
Llobregat, Barcelona, Spain
Received: September 14, 2021
Accepted: October 29, 2021
Published online: December 15, 2021
Correspondence to:
Javier Villacorta, javier.villacorta@salud.madrid.org
© 2021 S. Karger AG, Basel
karger@karger.com
www.karger.com/nef
DOI: 10.1159/000520727
Keywords
Glomerulonephritis · Antineutrophil cytoplasmic antibody ·
Vasculitis
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated
vasculitis (AAV), characterized by the presence of autoanti-
bodies to neutrophil cytoplasmic antigens, proteinase 3
(PR3), and myeloperoxidase (MPO), typically involves small
blood vessels of the respiratory tract and kidneys. It includes
distinct clinical syndromes: microscopic polyangiitis (MPA),
granulomatosis with polyangiitis (GPA), and eosinophilic
GPA. GPA is commonly associated with PR3-ANCA, while
MPA is associated with MPO-ANCA. AAVs have a complex
pathogenesis, influenced by genetics and environmental
factors. There is evidence for a loss of tolerance to neutrophil
proteins, which leads to ANCA-mediated neutrophil activa-
tion and injury, with effector T cells and activation of the al-
ternative pathway of the complement also involved. Ad-
vances in immunosuppressive treatment have drastically re-
duced mortality of AAV in the past decades, opting for a
more individualized approach. Careful assessment of ANCA
specificity, disease activity, organ damage, and quality of life
allows for a tailored immunosuppressive therapy. Contem-
porary AAV treatment is characterized by regimens that min-
imize the cumulative exposure to glucocorticoids and cyclo-
phosphamide, and novel approaches including comple-
ment blockage and immunosuppressant combinations
might be the standard of care in the future. In this review, we
examine the pathogenesis, clinical approach, and evidence-
based treatment options for the management of AAV pa-
tients. © 2021 S. Karger AG, Basel
Introduction
Antineutrophil cytoplasmic antibody (ANCA)-associ-
ated vasculitis (AAV) is a group of disorders character-
ized by inflammation and destruction of small- and me-
dium-sized blood vessels and the presence of circulating
ANCAs [1]. It comprises distinct clinical syndromes: mi-
croscopic polyangiitis (MPA), granulomatosis with poly-
angiitis (GPA), and eosinophilic GPA (EGPA) [2]. Sero-
logical classification of AAV into proteinase 3-ANCA
(PR3-ANCA) disease and myeloperoxidase-ANCA
(MPO-ANCA) disease is crucial [3]. GPA is commonly
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
122
DOI: 10.1159/000520727
associated with PR3-ANCA, while MPA is associated
with MPO-ANCA. However, differences in genetics,
pathogenesis, risk factors, treatment responses, and out-
comes associate closely with ANCA specificity (PR3 vs.
MPO-ANCA) than with clinical syndrome [4]. There-
fore, the ANCA serotype may guide clinical and thera-
peutic approaches, leading to a more personalized medi-
cine for AAV patients. Renal involvement is present in
the majority of patients with AAV, and the consequences
of a missed or delayed diagnosis of renal vasculitis are po-
tentially life-threatening. Advances in immunosuppres-
sive treatment have drastically reduced mortality of AAV
in the past decades with higher rates of remission ob-
served in recent trials [5]. However, infection rates are
still high, and current regimens aim to minimize the cu-
mulative exposure to glucocorticoids and cyclophospha-
mide (CYC). In this review, we discuss the pathogenesis,
clinical classification, diagnostic approach, potential bio-
markers, and evidence-based treatment regimens for pa-
tients with AAV.
Epidemiology
AAV is an infrequent disease with an incidence of
about 20 per million population per year in North Amer-
ica and Europe [6]. Incidence increases with age, with a
peak incidence during the seventh decade of life and a
marked predominance of male patients. AAV has a high-
er prevalence in Caucasic and Asian populations than Af-
rican-American populations [7]. There is notable geo-
graphic variation, with GPA being more common in
Northern Europe and Pacific area countries, whereas
MPA is more common in Southern Europe and Asia [8].
Infection with Staphylococcus aureus may trigger epi-
sodes of AAV since chronic nasal carriage of S. aureus has
been associated with an increased risk for disease relapse
[9]. Other environmental factors include silica exposure,
hydrocarbon exposure, pesticides, and medications such
as hydralazine and propylthiouracil [10–12].
Genetic Studies
Two genome-wide association studies in North Amer-
ican and European populations have identified disease
susceptibility loci in AAV [4]. GPA is associated with sin-
gle-nucleotide polymorphisms in HLA-DP, PRTN3 (en-
coding PR3), and SERPINA1 (encoding α1-antitrypsin, a
protease acting as the major inhibitor of PR3). PR3-AN-
CA disease-associated variants in PRTN3 and SERPINA1
support the hypothesis that PR3-ANCA is not merely an
epiphenomenon in AAV but plays a central role in the
pathogenesisofthisdisease.Bycontrast,MPAwasassoci-
atedwithHLA-DQpolymorphisms.Notably,thestrength
of these genetic associations was greater with respect to
ANCA specificity (PR3-ANCA or MPO-ANCA) than for
the clinical phenotype (GPA or MPA) [13]. These data
support for the concept that PR3-AAV and MPO-AAV
are distinct autoimmune syndromes.
Pathogenesis
AAV pathogenesis has classically been focused on the
activity and interactions of the neutrophil. They have a
double role as the target of the ANCAs and effectors of
the tissue damage after degranulation induced by these
antibodies. The exposure of MPO and PR3 in the surface
of the cellular membrane of the neutrophils is necessary
for the binding of the ANCA and requires a pre-activa-
tion process known as priming. Various microbial, en-
dogenous, or environmental agents and pro-inflammato-
ry cytokines such as interleukin (IL)-1, IL-18, and TNF-α
can prime neutrophils. Once primed, neutrophils achieve
their maximal phagocytic potential and response more
intensively to a second stimulus such as ANCA binding.
Proteolytic enzymes and free oxygen radicals are released
from neutrophil granules into the damage site. Neutro-
phils also release chemokines that recruit more neutro-
phils and other inflammatory cells [14]. Moreover, MPO
and PR3 can exert direct damage on endothelial cells [15].
In addition, neutrophils from AAV patients are more
prone to NETosis, spontaneously or induced by ANCA,
and tissular deposition of NETs also contributes to vas-
cular endothelial damage [16] (Fig. 1).
How ANCAs are produced is debated. MPO and PR3
are mostly circumscribed to the primary granules of the
neutrophil’s cytoplasm and are rapidly eliminated by spe-
cific inhibitors after neutrophil degranulation, thus stay-
ing out of reach of the immune system [17]. Several hy-
potheses have been drawn regarding how these proteins
can become immunogenic, such as the molecular mim-
icry with bacterial antigens hypothesis [18], the comple-
mentary peptide hypothesis [19], or the exposure of MPO
and PR3 as components of the NETs in NETosis process-
es [16].
In recent years, this neutrophil-focused view of the
AAV pathogenesis is being abandoned in favor of the
consideration of the disease as a multistep process with
ANCA Vasculitis: An Individualized
Approach
123
Nephron 2022;146:121–137
DOI: 10.1159/000520727
the involvement of other immune cells. In this line, ex-
tensive T-cell infiltrates have been identified in damaged
tissue from AAV patients, as well as forming part of the
characteristic granulomas [20]. Moreover, preclinical
models of AAV are attenuated in the absence of different
T-cell subsets, thus suggesting the importance of these
cells for the induction of the disease [21]. The ameliora-
tion of the disease after T-cell directed therapies is also a
proof of concept of the T-cell involvement [22]. The aber-
rant T-cell response in AAV consists on the polarization
toward predominant Th1 [23] and Th17 [24] phenotypes
during the acute phase of the disease, together with a nu-
merical and functional impairment in T regulatory lym-
phocytes [25]. Some authors described a higher propor-
tion of activated CD8+ T cells in AAV patients together
with a higher expression of IL-7 and T-cell receptor sig-
naling pathways in these cells [26]. Altered T-cell co-
stimulation has also been described in AAV, consisting of
a decreased CD28 expression and CTLA4 overexpression
with diminished inhibitory ability, thus resulting in resis-
tance to anergy and contributing to a more pronounced
pro-inflammatory state [27].
The participation of the alternate pathway of the
complement has been demonstrated in AAV. Despite
the paucity of the immune deposits in AAV lesions, el-
evated serum C5a levels have been described in active
AAV [28]. C5a acts as a powerful neutrophil priming
agent via the C5a receptor present in this cell. At the
same time, neutrophils release properdin that promotes
further generation of C5a [29]. Based on this rationale,
the utility of the C5a receptor inhibitor as induction to
remission treatment in active AAV has recently been
published [30].
Resting neutrophil
Plasma leakage and
fibrinoid necrosis
Endothelial damage
Chemotaxis of
inflammatory cells
Proteolytic enzymes release
NETosis
Direct ANCA damage
ANCA binding
Microbial, environmental or
endogenous agents,
proinflammatory cytokines,
C5a complement fraction
Primed neutrophil
ROS release
Molecular mimicry with bacterial antigens
Immune response to complementary
peptides
Exposure of MPO and PR3 after netosis
ANCA production
Fig. 1. Pathophysiology. MPO and PR3 are inside cytoplasmic
granules in resting neutrophils. Pre-activation (priming) of the
neutrophils by environmental, infectious, or pro-inflammatory
agents is required to expose MPO and PR3 in the cell surface. Once
exposed, ANCAs can bind to MPO and PR3. ANCA binding leads
to neutrophil degranulation that releases ROS and proteolytic en-
zymes that cause endothelial damage. The rupture of the endothe-
lium allows plasma leakage that causes fibrinoid necrosis. MPO,
myeloperoxidase; PR3, proteinase 3; ANCA, antineutrophil cyto-
plasmic antibody; ROS, reactive oxygen species.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
124
DOI: 10.1159/000520727
Clinical Presentation
AAV is a heterogeneous group of autoimmune multi-
system conditions that can present with a wide variety of
signs and symptoms. Clinical manifestations depend on
the size, number, and type of affected vessels and the or-
gans involved, disease stage, and activity [5]. In addition
to organ-specific manifestations, patients often present
with constitutional symptoms such as fever, malaise,
weight loss, anorexia, myalgias, and arthralgias which re-
late to the systemic autoimmune pathophysiology [1].
The upper respiratory tract is usually compromised of de-
structive inflammatory lesions causing nasal, sinus, tra-
cheal, and/or ear abnormalities [31]. Upper respiratory
tract and ear-nose-throat (ENT) involvement frequently
is manifested by recurrent epistaxis, mucosal ulceration,
nasal septum deformities and perforation, sinusitis, otitis
media, or subglottic stenosis [32]. Pulmonary manifesta-
tions of AAV include pulmonary nodules and diffuse al-
veolar hemorrhage. Alveolar hemorrhage occurs as a re-
sult of pulmonary capillaritis and presents with hemop-
tysis and dyspnea. The most reported ocular involvement
in AAV includes orbital disease and scleritis [33]. Cuta-
neous [34], neurological [35], or enteric [36] involvement
is also frequent (Table 1).
Regarding to kidney involvement in AAV, it manifests
as a rapidly progressive glomerulonephritis. Usually, it is
accompanied by a nephritic syndrome with hematuria
and proteinuria. Less frequently, renal involvement in
AAV presents as subacute or chronic nephritis. Renal
AAV can lead to end-stage renal disease.
The different AAV disease phenotypes are composed of
adifferentialpatternofassociationsoforgandamage.Prev-
alence of renal involvement ranges between 75 and 90%
and is higher in MPA than inGPA or EGPA [1]. Necrotiz-
ing glomerulonephritis is present in >80% patients diag-
nosed with MPA and up to 50% patients present with pul-
monaryinvolvement.ThirtypercentofMPApatientspres-
ent with neurological, skin, or gastrointestinal symptoms.
In GPA patients, necrotizing vasculitis is accompanied by
granulomatous inflammation. ENT and upper airway in-
volvement are more frequent in GPA patients than in MPA
patients. EGPA presents on a background of eosinophilia
and asthma, and sinuses abnormalities are commonly as-
sociated. Around one-third of patients have renal manifes-
tations. Some patients with GPA or MPA present with vas-
culitis limited to a single organ, such as the kidneys or
lungs, which may represent the early stages of AAV. How-
ever, in MPO-ANCA+ patients with MPA, isolated renal
disease or isolated pulmonary fibrosis is not infrequent.
Classification Criteria and Diagnosis
The classification of the systemic vasculitis has been
controversial for several decades. The American College
of Rheumatology Criteria (ACR) was published in 1990
and included only GPA and EGPA but not MPA [37].
Lately, the Chapel Hill Consensus Conference (CHCC)
definitions developed in 1994 and revised in 2012 added
the knowledge of the etiopathogenesis of vasculitis. A
new tree hierarchy was developed which recognized that
some conditions cannot be simply classified by vessel size
and suggested the use of surrogate markers of the disease,
such as ANCAs. AAV was recognized as a specific type of
small-vessel vasculitis along with immune complex-me-
diated vasculitis (Fig. 2) [2].
In clinical practice, AAV serological diagnosis is based
on the finding of ANCA positivity in sera from patients
with consistent signs and symptoms. The presence of
ANCA can be evaluated by means of the indirect immuno-
fluorescence(IIF)testonhumanethanol-fixedneutrophils
[38] by enzyme-linked immunoassay and more recently by
chemiluminescent immunoassay. C-ANCA describes an
IIF pattern consisting on a diffuse granular cytoplasmic
staining predominantly associated to the presence of auto-
antibodies targeting PR3. On the contrary, the IIF pattern
P-ANCA consists on a perinuclear staining related to the
presence of autoantibodies against MPO [39]. Results re-
Table 1. Serological and clinical features of AAV
GPA, % MPA, % EGPA, %
PR3/C-ANCA 80–90 10–20 10–30
MPO/P-ANCA 10–15 70–80 40–60
Negative ANCA 5 10–20 10–50
Cutaneous 40 40 50
Kidney 80 90 40
Pulmonary 90 50 70
ENT 90 30 50
Neurological 50 30 70
Gastrointestinal 30–50 30–50 30–50
Musculoskeletal 60 60 50
Eyes 30 20 10
ANCA, antineutrophil cytoplasmic antibody; PR3, proteinase 3;
MPO, myeloperoxidase; GPA, granulomatosis with polyangiitis;
MPA, microscopic polyangiitis; EGPA, eosinophilic granulomatosis
with polyangiitis; RLV, renal-limited vasculitis; ORL, otorhinolaryn-
gological; AAV, antineutrophil cytoplasmic antibody-associated
vasculitis; ENT, ear, nose, and throat.
ANCA Vasculitis: An Individualized
Approach
125
Nephron 2022;146:121–137
DOI: 10.1159/000520727
trieved from the IIF test, as a screening test, should be con-
firmed by enzyme-linked immunoassay [40]. In the recent
years, chemiluminescent immunoassay has been devel-
oped in order to improve sensitivity and specificity in the
detection and titration of ANCAs [39].
Regarding the clinical syndrome, MPA-AAV patients
often show a P-ANCA pattern associated to the presence
of anti-MPO antibodies [41], but they can also exhibit
anti-PR3 antibodies. The IIF pattern most commonly as-
sociated with GPA, but not exclusive, is C-ANCA, related
to anti-PR3 antibodies [42]. In EGPA, <50% of patients
have detectable ANCAs, more frequently anti-MPO an-
tibodies [43].
More recently, results from epidemiological outcomes
and genetic studies in AAV suggest that patients should be
classified by ANCA specificity rather by the clinical syn-
drome [4]. The new Diagnostic and Classification Criteria
of Vasculitis Study aims to provide new validated classifi-
cation and diagnostic criteria for systemic vasculitis [44].
Disease Activity and Damage Scores
Validated tools to assess disease activity are Birming-
ham Vasculitis Activity Score (BVAS) and the Five-Fac-
tor Score. The BVAS includes 10 categories of new or
worsening symptoms presented <4 weeks until the re-
cord. A BVAS score of 0 represents remission, ≥1 repre-
sents active disease, and refractory disease is active dis-
ease despite treatment [45]. The 2009 Five-Factor Score
for EGPA comprises serum creatinine, age (>65 years),
cardiomyopathy, gastrointestinal involvement, and the
absence of ENT manifestations [46]. On the other hand,
the Vasculitis Damage Index is used to assess chronic
damage from the disease and treatment and includes
musculoskeletal, skin, and mucous membranes and ocu-
lar, ENT, pulmonary, cardiovascular, the peripheral vas-
culature, gastrointestinal, renal, and neuropsychiatric
systems, with an eleventh category for other systems [47].
Biomarkers
There is a rising interest in the use of biomarkers in
screening disease, diagnosing, staging, monitoring thera-
peutic interventions, predicting outcomes or adverse
events, or identifying cell types in AAV. Although con-
troversial, the most accepted classical biomarker of active
AAV is the ANCA titer. In a subgroup of 104 AAV pa-
tients with renal involvement, Kemna et al. [48] found a
correlation between ANCA titer rise and disease relapse.
Lionaki et al. [49] described that ANCA-PR3 specificity
Large vessel
Takayasu arteritis
Giant cell arteritis
Medium vessel
Polyarteritis nodosa
Kawasaki disease
Cryoglobulinaemic vasculitis
lgA vasculitis
Hypocomplementemic urticarial vasculitis
Anti-GBM disease
ANCA-associated vasculitis
Small vessel
Fig. 2. Classification of AAV. AAV, antineutrophil cytoplasmic antibody-associated vasculitis; Ig, immunoglobulin.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
126
DOI: 10.1159/000520727
predicts relapse in AAV patients with kidney involve-
ment, conferring almost twice the relapse risk compared
to ANCA-MPO vasculitis.
Based on the disease pathophysiology, neutrophil ac-
tivation biomarkers are of special interest in AAV. After
ANCA binding and full activation of these cells, neutro-
phil microparticles and NETs are released and can be
measured in the serum of AAV patients for the assess-
ment of disease activity [50, 51]. Calprotectin is contained
in the cytoplasmic granules of neutrophils and released
after degranulation in active AAV. Calprotectin binds en-
dothelial cells and amplifies endothelial damage and in-
flammation, by increasing IL8, ICAM1, and leukocyte re-
cruitment [52]. Pepper et al. [52] demonstrated calpro-
tectin expression by immunohistochemistry in kidney
biopsies from AAV patients localized in crescents and ar-
eas of endocapillary proliferation. They also found that
serum calprotectin is elevated in patients in the acute
phase of AAV compared to patients in remission [53].
Our group described higher serum calprotectin levels in
remission in those patients with worse renal outcomes
during the follow-up and found higher urinary calprotec-
tin levels in the acute patients than in the remission pa-
tients [54]. Also, in the field of urinary biomarkers,
suCD163 has shown promising results. suCD163 is pro-
duced by macrophages composing the inflammatory in-
filtrates in AAV, released to the urinary space, and mea-
sured in the urine. Higher suCD163 levels have been no-
ticedinacuteAAVwithrenalinvolvementthanremission
[55]. In a prospective study, suCD163 consistently in-
creased compared to previous levels during the follow-up
of patients in remission who experienced relapse [56].
suCD163 has been evaluated in combination with sCD25
[57] or MCP1 [58], showing an improved performance as
a disease activity biomarker.
Due to their emerging role in disease pathogenesis,
some T-cell subpopulations or byproducts have been de-
scribed as AAV biomarkers. Our group found higher fre-
quencies of MPO/PR3-specific Th17 cells in acute AAV
than remission and suggested higher specific Th17 count
in remission as a surrogate marker of subclinical activity
[59]. Morgan et al. [60] found that patients with a higher
Treg count at presentation entered earlier in remission
than those with a lower Treg count and a negative corre-
lation between Tregs and the disease relapse rate. In the
same line, Yoshimura et al. [61] correlated a lower FOXP3
staining – a key transcription factor for the suppressive
ability of Tregs – in AAV renal biopsies with the require-
ment of maintenance. McKinney et al. [26] found that the
higher expression of a particular subgroup of genes re-
lated to T-cell survival, and expansion of memory CD8 T
cells was strongly associated with a shorter time to relapse
after induction to remission treatment and identified
those individuals with a greater propensity for relapse, al-
lowing the customization of the therapy.
Inthesettingoftherisinginterestinthealternatepath-
way of the complement as a therapeutic target in AAV,
complement fractions have also been studied as biomark-
ers. Villacorta et al. observed that serum C3 at baseline
was predictive of renal and global survival in AAV pa-
tients, with the worse outcomes associated with C3 con-
sumption [62] and that C3d-positive staining in diagnos-
tic kidney biopsy was associated with the severity of renal
impairment and with a lower response rate to treatment
[63].
Treatment
Remission Induction Therapy
Advances in immunosuppressive treatment have dras-
tically reduced mortality of AAV in the past decades. Re-
mission rates obtained in different clinical trials range be-
tween 53 and 90% [64] (Table  2). However, mortality
rates despite immunosuppressive treatment are not neg-
ligible, especially in those patients presenting with severe
organ involvement, such as renal failure or alveolar hem-
orrhage [65]. Infection constitutes the major cause of
death within the first year after the onset, followed by ac-
tive vasculitis damage. Therefore, AAV therapy should
try to control disease activity but minimize side-effect de-
velopment, mainly infections.
The goal of induction therapy is to achieve remission
within the first 3 months, by employing more intensive
immunosuppression. Before initiation of therapy, careful
assessment of disease activity, organ damage, and quality
of life is crucial and may guide the use of potentially tox-
ic therapies. Remission has been defined as improvement
or stability of renal function, resolution of hematuria, and
the absence of extrarenal features of disease activity.
Treatment should be initiated as soon as AAV with organ
and life-threatening manifestations are suspected, even in
the absence of histological confirmation of vasculitis
since a delay in treatment can lead to worse outcomes.
CYC-Based Remission Induction Treatment
AAV with organ and life-threatening manifestations
has been classically treated with combination therapy of
glucocorticoids and CYC for 3–6 months. CYC can be
given orally or intravenously. Both routes of administra-
ANCA Vasculitis: An Individualized
Approach
127
Nephron 2022;146:121–137
DOI: 10.1159/000520727
Table
2.
Induction
immunosuppressive
protocol
according
to
different
trials
Study
Drug
comparison
Induction
immunosuppressive
protocol
Maintenance
therapy
Results
immunosuppressive
drug
doses
and
duration
corticosteroid
dose
corticosteroid
tapering
CYCLOPS
RCT
(2009)
ivCYC
versus
oral
CYC
ivCYC
15
mg/kg
every
2–3
weeks
for
3–6
months
Oral
prednisolone
1
mg/kg
Tapering
to
12.5
mg/
day
at
month
3
and
to
5
mg
at
month
18
Oral
AZA
2
mg/kg/day
since
month
3–6
to
month
18
No
differences
in
remission
rates.
Less
cumulative
dose
in
the
ivCYC
group
Oral
CYC
2
mg/kg/day
for
3–6
months
RAVE
RCT
(2010)
RTX
versus
oral
CYC
RTX
375
mg/m
2
/week
for
4
weeks
1–3
pulses
1
g
iv
methylprednisolone,
followed
by
oral
prednisolone
at
1
mg/kg/day
Tapering
to
zero
at
month
5
Placebo
maintenance
RTX
noninferior
to
oral
CYC
for
induction
remission
RTX
superior
for
relapsing
vasculitis
or
PR3-positive
patients
Oral
CYC
2
mg/kg/day
for
3–6
months
Oral
AZA
2
mg/kg/day
since
month
3–6
to
month
18
RITUXVAS
RCT
(2010)
RTX
versus
ivCYC
RTX
+
2
doses
ivCYC±PEX/
SDS
RTX
375
mg/m
2
/week
for
4
weeks
+
CYC
15
mg/kg
with
the
first
and
third
RTX
1
pulse
1
g
iv
methylprednisolone,
followed
by
oral
prednisolone
at
1
mg/kg/day
Tapering
to
5
mg/day
at
month
6
Withdrawn
at
month
12
No
maintenance
RTX
noninferior
to
ivCYC
for
induction
remission
ivCYC
15
mg/kg
every
2
weeks
for
3–6
months
Oral
AZA
2
mg/kg/day
since
month
3–6
to
month
24
Pepper
et
al.
[86]
Cohort
study
(2019)
LDS
versus
SDS
from
historic
cohort
of
EUVAS
trials
RTX
+
ivCYC
RTX
1
g
at
day
0
and
day
7
CYC
500
mg,
6
doses
every
2
weeks
2
pulses
250–500
mg
methylprednisolone
at
days
0
and
7,
followed
by
0.5
mg/kg/day
(max
30
mg)
days
2–6
Withdrawn
at
day
7
(maximum
duration
3
weeks)
Oral
AZA
2
mg/kg/day
No
differences
in
remission
rates
RTX
1
g
at
day
0
and
day
14
CYC
500
mg,
6
doses
every
2
weeks
1
pulse
250–1,000
mg
methylprednisolone,
followed
by
60
mg/day
1
week,
followed
by
45
mg/day
1
week
Withdrawn
at
week
2
(5
patients
received
>30
days
of
prednisolone)
MYCYC
RCT
(2019)
Oral
MMF
versus
ivCYC
Oral
MMF
2
g/day
until
remission
Oral
prednisolone
1
mg/kg
Tapering
to
5
mg/day
at
month
6
Prednisolone
5
mg/day
+
oral
AZA
2
mg/kg/day
since
remission
to
month
18
No
differences
in
remission
rates
At
18
months:
more
relapses
with
MMF,
especially
in
PR3-
ANCA
patients
IvCYC
15
mg/kg
every
2–3
weeks
until
remission
CYCLOWVAS
Cohort
study
(2019)
LDS
versus
SDS
from
historic
cohort
of
EUVAS
trials
RTX
+
ivCYC
RTX
1
g
at
day
0
and
day
14
CYC
10
mg/kg
to
maximum
of
500
mg,
6
doses
every
2
weeks
Oral
prednisolone
1
mg/kg
(maximum
dose
60
mg)
Tapering
to
10
mg/day
at
week
13
Oral
AZA
2
mg/kg/day
since
month
3
to
month
18–24
High
rate
remission
Reduced
risk
of
death,
ESRD
and
relapses
compared
with
the
historic
cohort
PEXIVAS
RCT
(2020)
LDS
versus
SDS
RTX/ivCYC±PEX
RTX/ivCYC
according
to
local
practitioner
PEX
according
to
the
PEXIVAS
trial
randomization
1–3
pulses
1
g
iv
methylprednisolone,
followed
by
oral
prednisolone
at
1
mg/kg/day
Tapering
to
5
mg/day
at
week
23–52
No
maintenance
specified
No
differences
in
remission
rates
between
groups
1–3
pulses
1
g
methylprednisolone,
followed
by
oral
prednisolone
at
1
mg/kg/day
Tapering
to
5
mg/day
at
week
15–16
ADVOCATE
RCT
(2021)
Avacopan
versus
prednisone
RTX
+
ivCYC
RTX
(375 mg/m
2
weekly
for
4
weeks)
or
CYC
orally
(2
mg/
kg
daily
for
14
weeks)
or
IV
(15
mg/kg
every
2–3
weeks
for
13
weeks
Prednisone:
60 mg
versus
avacopan:
30 mg
orally
twice
daily
for
52
weeks
without
prednisone
Taper
over
21
weeks
Oral
AZA
2
mg/kg/day
since
month
3–6
to
month
18
after
CYC
The
RTX
arm
did
not
receive
maintenance
At
weeks
26
and
52:
Noninferior
remission
rate
in
the
avacopan
group
compared
to
prednisone
CYC,
cyclophosphamide;
ivCYC,
intravenous
cyclophosphamide;
AZA,
azathioprine;
RTX,
rituximab;
MMF,
mycophenolate
mofetil;
PEX,
plasma
exchange;
SDS,
standard
dose
of
steroids;
LDS,
low
dose
of
steroids;
EUVAS,
European
Renal
Association
and
the
European
Vasculitis
Society.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
128
DOI: 10.1159/000520727
tion were evaluated in the CYCLOPS trial, in which pa-
tients with a new diagnosis of AAV were randomly as-
signed to receive either oral CYC or intravenous CYC
[66]. There was no difference in remission rates between
the 2 groups (88.1% vs. 87.7%), but the cumulative dose
was significantly higher in the oral CYC group (15.2 vs.
8.2 g) and was associated with a higher incidence of leu-
kopenia. Long-term follow-up of the trial showed that al-
though the oral CYC group was at lower risk for relapse,
this did not translate into differences in renal or overall
survival [67]. For this reason, several clinical guidelines
recommend intravenous CYC as induction therapy [67,
68].
CYC is associated with several serious adverse effects
such as infection, bone marrow suppression, hemorrhag-
ic cystitis, and infertility [68]. Several studies have linked
CYC therapy with an increased incidence of malignances,
mainly skin cancer, myeloid malignancies, and bladder
cancer [69, 70]. This increased risk for the development
of tumors is especially evident when the cumulative dose
is >20 gr [71]. Therefore, induction treatment with CYC
should be avoided in patients with cancer history, those
with high risk or active infection, and young patients with
gestational desire.
Rituximab-Based Remission Induction Treatment
Given the substantial toxicity associated with cumula-
tive CYC use and the relapsing nature of AAV, the use of
rituximab (RTX), a chimeric anti-CD20 monoclonal an-
tibody, has been widely employed as induction therapy
[71]. Two randomized controlled trials, RAVE and
RITUXVAS, evaluated the efficacy of RTX for remission
induction in GPA and MPA [72, 73]. In the RAVE trial,
mainly composed by PR3-ANCA patients with both new
and relapsing diseases, but without severe renal failure,
the RTX arm received methylprednisolone pulses, and
the prednisone dosage was tapered to zero within 6
months. On the other hand, the RITUXVAS trial enrolled
only patients with newly diagnosed vasculitis with more
severe kidney disease, including patients requiring dialy-
sis (20%). In contrast to the RAVE trial, the RTX arm re-
ceived 2 doses of intravenous CYC, and the use of plas-
mapheresis was allowed. Both studies demonstrated that
RTX therapy was noninferior to CYC for remission in-
duction, with comparable rates of mortality and adverse
events. A subanalysis of the RAVE trial performed among
patients with relapsing disease concluded that RTX was
superior to CYC in PR3-ANCA patients with relapsing
disease since a higher rate of remission in the RTX arm
was observed [74]. Based on this, current guidelines rec-
ommend RTX as first-line treatment for PR3-ANCA pa-
tients, relapsing disease, refractory disease, and those
with contraindications to CYC [75, 76] (Fig. 3).
Specific measures such as prevention of hepatitis B vi-
rus reactivation and hypogammaglobulinemia surveil-
lance must be taken when receiving RTX therapy [77]. Up
to 25% of patients with AAV present hypogammaglobu-
linemia at the onset of AAV disease and after RTX ther-
apy low levels of immunoglobulin (Ig) G are observed in
up to 50–60% of patients [78]. In most cases, hypogam-
maglobulinemia is mild and transient, and IgG levels re-
turntonormalwithoutspecificmeasureswithin6months
after RTX infusion. However, severe hypogammaglobu-
linemia occurs in 4.2% of patients and contributes to re-
current infections, requiring the use of intravenous Ig.
Therefore, Ig levels should be checked at baseline and at
least before each RTX infusion [79].
Individuals with chronic hepatitis B and previously in-
fected but serologically cleared HBV infection are both
susceptible to HBV reactivation after RTX therapy [80].
Identification of patients at risk and institution of pro-
phylactic antiviral therapy prior to initiation of RTX is
essential [81]. Finally, late-onset neutropenia is a rare side
effect of RTX observed among AAV patients that mani-
fests as abrupt and severe neutropenia within 2–6 months
following the last dose of RTX and may require filgrastim
administration in some cases [82].
Mycophenolate-Based Remission Induction Treatment
The use of mycophenolate mofetil (MMF) for induc-
tion remission therapy was evaluated in a randomized
controlled trial (MYCYC) which compared intravenous
CYC versus oral MMF (2–3 g/d) for treatment of newly
diagnosed AAV patients without severe kidney failure
[83]. The remission rates observed at 6 months were com-
parable between both groups (67% vs. 61%). However,
relapses were more common in the MMF group, mostly
in PR3-ANCA-positive patients. Therefore, the study
suggests that MMF and glucocorticoids can be used as a
first-line induction therapy, mainly in patients with
MPO-ANCA who have mild to moderate renal involve-
ment without life-threatening extrarenal vasculitis.
Novel Strategies for Remission Induction Therapy:
Steroid Minimization Regimens
Classically, oral glucocorticoids have constituted a
cornerstone of AAV induction therapy due to its rapid
effect which allows disease control in the meanwhile oth-
er immunosuppressives to have an effect. However, glu-
cocorticoids have an extensive side-effect profile, includ-
ANCA Vasculitis: An Individualized
Approach
129
Nephron 2022;146:121–137
DOI: 10.1159/000520727
ing hypertension, hyperglycemia, weight gain, bone dis-
ease, psychiatric disorders, gastrointestinal bleeding, and
long-term risks for cardiovascular disease [84].
Although intravenous pulse methylprednisolone at
the onset of therapy for severe disease is conventionally
administered, its benefits have not been adequately stud-
ied. In recent years, several studies have explored the em-
ploy of lower dose regimens of steroids for induction
therapy and similar efficacy with a reduced risk of severe
infections was observed. The CYCLOWVAS study pro-
posed a combination of RTX with quinquennial low-dose
pulses of CYC associated with a rapid taper of glucocor-
ticoids within the first month [85]. A high remission rate
was observed in this study and when compared with an
European Renal Association and the European Vasculitis
Society (EUVAS) historic cohort, a reduction of the risk
of progression to end-stage renal disease and death was
shown. Another observational study employing this im-
munosuppression protocol represented a step forward
toward glucocorticoid withdrawal [86]. In this study,
prednisone was discontinued within the first 2 weeks of
therapy. Patients with severe renal failure were not ex-
cluded, and however, the remission rates observed were
very high (>90%). The rapid withdrawal of prednisone
was associated with a significant lower incidence of car-
diovascular events and diabetes. However, further trials
are needed to confirm the safety and superiority of this
combination regimen. Finally, the recent PEXIVAS trial
demonstrated that a reduction of the prednisone dose to
20 mg daily by 7 weeks and 5 mg daily by 20 weeks was as
Mild to moderate kidney failure
New onset
MPO PR3/MPO
Relapsed disease
Severe kidney failure
New onset/relapsed disease
PR3/MPO
RTX + SDS a)
RTX + ivCYC + LDS (*) e) RTX + ivCYC + SDS g)
RTX + ivCYC + LDS (*) h)
PR3
RTX + SDS a)/LDS d)
No remission Remission No remission No remission No remission
Consider stop
treatment and/or
dialysis
Swich therapy
Remission Remission
Maintenance therapy
RTX 500 mg every 4–6 months i) (+)
AZA j)
ivCYC ivCYC
RTX + SDS a)/LDS d)
ivCYC + SDS b)/LDS d)
MMF + SDS c)
RTX + iv CYC + LDS (*) e)
Avacopan + RTX/ivCYC f)
Fig. 3. Treatment algorithm for AAV. Mild to moderate kidney
failure: GFR >15 mL/min/1.73 m2
; severe kidney failure: GFR <15
mL/min/1.73 m2
. a) RAVE RCT protocol; b) CYCLOPS RCT pro-
tocol; c) MYCYC RCT protocol; d) PEXIVAS trial; e) CYCLOW-
VAS case-control trial protocol (RTX + quinquennial low-dose
CYC); f) AVACOPAN RCT protocol; g) RITUXVAS RCT proto-
col (RTX + quinquennial low-dose CYC); h) case control study
protocol from Pepper et al. [86]; i) MAINRITSAN 1 RCT protocol;
j) CYCAZREM RCT protocol. (*) Patients with intolerance or
contraindication to corticosteroids (+) or according to ANCA sta-
tus or CD19 B-cell repopulation (MAINRITSAN 2 protocol).
Maintenance for 24 months in patients with low risk of relapse and
up to 48 months in PR3-ANCA patients and those with high risk
for relapsing. PR3, proteinase 3; MPO, myeloperoxidase; RTX,
rituximab; SDS, standard dose of steroids; LDS, low dose of ste-
roids; ivCYC, intravenous cyclophosphamide; MMF, mycopheno-
late mofetil; AZA, azathioprine; RCT, randomized clinical trial;
AAV, antineutrophil cytoplasmic antibody-associated vasculitis;
CYC, cyclophosphamide.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
130
DOI: 10.1159/000520727
effective as a higher dose regimen and implied a substan-
tial reduction in the infection rates [87].
Complement Blockage: Betting on the Future
Given the knowledge of the crucial role of complement
activation in the pathogenesis of AAV, therapies target-
ing the alternative pathway and particularly the anaphyl-
atoxin C5a have emerged [28]. The ADVOCATE trial
was a phase 3 study which analyzed standard immuno-
suppression with CYC or RTX combined with a selective
C5a receptor inhibitor avacopan or oral prednisone on a
tapering schedule [30]. At week 26, the proportion of pa-
tients in disease remission (BVAS = 0) in the avacopan
group was comparable to the prednisone group (72.3%
vs. 70.1%) [29]. Sustained remission at week 52 was ob-
served in 65.7% of the patients receiving avacopan and in
54.9% receiving prednisone. Therefore, avacopan nonin-
feriority was met at weeks 26 and 52 (p < 0.001), and su-
periority was met at week 52 only (p = 0.007). Avacopan
patients reported better health-related quality of life in
several measures (physical health, health perception,
emotional health, and vitality).
However, the study had some limitations. First, the
large number of patients in the avacopan arm (86%) who
received nonstudy-supplied glucocorticoids within the
first 6 months may have had an impact on the study drug
effect. Second, the lack of maintenance therapy in the
RTX subgroup may also have had some positive impact
outcome for the avacopan arm since the difference for
long-term disease remission was only observed among
patients treated with RTX. Finally, similar infection rates
were observed in both groups of treatment, but a greater
proportion of avacopan-treated patients had adverse
events associated with hepatic abnormalities. Despite
these concerns, the avacopan groups were noninferior to
the standard prednisone group in the study, highlighting
a potential approach in which glucocorticoid doses can be
minimized. Another phase II study is currently analyzing
the employ of vilobelimab (IFX-1), a first-in-class anti-
C5a antibody, added to standard of care and will certain-
ly provide further information about complement inhibi-
tion in AAV induction therapy [88].
Plasma Exchange: Unlearning What Has Been
Learned?
The benefit for considering plasma exchange (PEX) in
treatment of patients AAV is that the removal of ANCAs
and other inflammatory mediators could promote earlier
reversal of the immunological response and minimize tis-
sue damage. However, PEX is an invasive therapeutic
method which could increase the risk of infection, hem-
orrhage, and catheter-related complications and when
frozen plasma is employed, transfusion-related adverse
reactions [89]. The MEPEX trial showed that adjuvant
therapywithPEXaddedtoCYCinAAVpatientspresent-
ing with severe kidney failure was associated with a 24%
risk reduction in progression to end-stage kidney disease
[90]. However, the longer term follow-up data showed no
difference in mortality or end-stage kidney disease [91].
The PEXIVAS enrolled a total of 704 MPO and PR3-AAV
patients with renal involvement and with or without pul-
monary compromise [87]. Treatment with PEX associ-
ated to standard immunosuppression (RTX or CYC) was
compared with no PEX, and a standard dose oral gluco-
corticoid regimen was compared with a reduced dose reg-
imen. Notably, serious adverse events, including serious
infections, were comparable among patients treated with
and without PEX but were less frequent among patients
receiving a low dose of steroids. After a median follow-up
of 3 years, the study showed no difference in primary end
points (death from any cause or kidney failure) between
groups receiving or not PEX, and subgroup analysis failed
to show a benefit among those patients requiring dialysis
at the onset (20%).
However, similarly to the MEPEX study, the analysis
at the 1-year follow-up showed a nonsignificant advan-
tage of PEX (HR, 0.77; 95% CI, 0.56–1.06), whereas sta-
tistical assessment at 3 and 6 months was not provided
despite short-term clear separation between survival
curves. Therefore, many nephrologists consider that a
short-term benefit of PEX leading to a free dialysis period
within the first year, as well as the absence of serious tech-
nique-related complications in the study, still justifies
PEX employ among patients presenting with severe renal
failure.
In the PEXIVAS trial, 61 (9%) patients with severe pul-
monary hemorrhage, defined as oxygen saturation ≤85%
or requirement of mechanical ventilation, were included.
Although PEX did not show a statistically significant ef-
fect on the primary outcome in these patients, a trend
toward the beneficial effect of PEX could be observed
(HR,0.67;95%CI,0.28–1.6).However,theimpactofPEX
on mortality alone was not provided, and the study had
not powerful enough to evaluate the true effect of PEX in
these patients.
Maintenance Therapy: Which One and for How Long?
AAV constitutes a chronic disease with a relapsing
course. Up to 30–60% of patients will present a relapse in
the course of the disease, mainly within the first 2 years
ANCA Vasculitis: An Individualized
Approach
131
Nephron 2022;146:121–137
DOI: 10.1159/000520727
Table
3.
Maintenance
immunosuppressive
protocol
according
to
different
trials
Study
Immunosuppressive
drug
induction
Maintenance
comparison
Maintenance
drug
Doses
and
duration
Steroid
maintenance
Results
CYCAZAREM
RCT
(2003)
Oral
CYC
+
SDS
Oral
CYC
versus
AZA
Oral
CYC
1.5
mg/kg/day
until
month
12,
followed
by
AZA
1.5
mg/kg/day
Prednisolone
10
mg/day
until
month
12,
then
tapering
to
7.5
mg/day
No
differences
in
relapse
rates
AZA
2
mg/kg/day
until
month
12,
then
tapering
to
1.5
mg/kg/day
IMPROVE
RCT
(2010)
CYC
+
SDS
MMF
versus
AZA
MMF
2
g/kg/day,
tapering
to
1.5
g/kg/day
after
12
months,
1
g/kg/day
after
18
months,
withdrawn
after
42
months
Prednisolone
15
mg/day
at
the
start
of
maintenance,
tapering
to
5
mg/day
after
12
months,
withdrawn
after
24
months
Higher
relapse
rates
in
the
MMF
group
AZA
2
mg/kg/day,
tapering
to
1.5
mg/kg/day
after
12
months,
1
mg/kg/day
after
18
months,
withdrawn
after
42
months
MAINRITSAN
RCT
(2014)
ivCYC
+
SDS
RTX
versus
AZA
RTX
500
mg
at
days
0
and
14,
then
at
months
6,
12,
and
18
Prednisone
tapering
to
5
mg/day
until
18
months,
then
withdrawn
according
to
physician
criteria
Greater
sustained
remission
in
the
RTX
group
AZA
2
mg/kg/day
for
12
months,
1.5
mg/kg/
day
for
6
months,
1
mg/kg/day
for
4
months
REMAIN
RCT
(2017)
CYC
+
SDS
AZA
+
LDS
for
24
months
versus
AZA
+
LDS
for
48
months
AZA
+
LDS
for
24
months
Withdrawn
AZA
at
month
24
Withdrawn
steroids
at
month
24
Greater
sustained
remission
in
the
prolonged
AZA
group
AZA
+
LDS
for
48
months
AZA
1
mg/kg/day
until
month
48
Tapering
prednisolone
from
5
to
7.5
mg/day
to
zero
at
month
48
MAINRITSAN
2
RCT
(2018)
CYC
+
SDS
Fixed
RTX
dosing
versus
tailored
RTX
dosing
Fixed
500 mg
IV
at
days
0
and
14
and
then
at
6,
12,
and
18
months
No
significant
difference
in
the
relapse
rate.
Less
RTX
infusions
in
the
tailored
group
Individualized
500 mg
IV
at
randomization
and
then
reinfusion
only
if
reappearance
of
CD19
or
ANCA
or
increased
titer
of
ANCA
Prednisone
tapering
to
5
mg/day
until
18
months,
then
withdrawn
according
to
physician
criteria
MAINRITSAN
3
RCT
(2020)
CYF/MTX/RTX
+
SDS*
RXT
for
18
months
(5
doses)
versus
RTX
for
36
months
(9
doses)
RTX
18
months
500
mg
at
day
0
and
14,
then
at
months
6,
12,
and
18
Prednisone
tapering
to
5
mg/day
until
18
months,
then
withdrawn
according
to
physician
criteria
Greater
sustained
remission
in
the
prolonged
RTX
group
RTX
46
months
500
mg
at
days
0
and
14,
then
at
months
6,
12,
and
18,
then
at
months
28,
34,
40,
and
46
CYC,
cyclophosphamide;
AZA,
azathioprine;
RTX,
rituximab;
MMF,
mycophenolate
mofetil;
MTX,
methotrexate;
SDS,
standard
dose
of
steroids;
LDS,
low
dose
of
steroids.
*Patients
in
the
MAINRITSAN
2
trial.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
132
DOI: 10.1159/000520727
after the withdrawal of immunosuppression. The appear-
ance of relapses implies a substantial increase in the mor-
bidity in these patients. Risk factors for relapse include
PR3-ANCA serotype, GPA phenotype, ENT involve-
ment, persistent ANCA-positive titers, and persistent mi-
crohematuria [86, 92].
The use of maintenance immunosuppression for re-
lapse prevention following successful remission induc-
tion is standard (Table 3). The first study to compare 2
maintenance regimens was the CYCAZAREM trial, in
which patients with AAV who achieved remission with
oral CYC were randomly assigned at 6 months to con-
tinue CYC therapy or switch to azathioprine (AZA) [93].
At 18 months, relapse rates were similar between both
arms (15.5 vs. 13.7%) but with less side effects in the AZA
group. The IMPROVE trial compared MMF to AZA and
showed higher rates of relapse in patients treated with
MMF. Therefore, MMF may be considered for remission
maintenance in patients who have intolerance to AZA.
Subsequently, the employ of RTX as maintenance
therapy showed dramatically lower rates of relapse. In the
MAINRITSAN 1 trial, patients who achieved remission
after CYC induction regimen were randomized to receive
either AZA or biannual semestral RTX infusions (500
mg) as maintenance therapy [94]. In the RTX arm, only
5% of treated patients experienced a major relapse com-
pared to 29% in the AZA group. This beneficial effect of
RTX in the prevention of relapses was independent of the
ANCA serotype, and no difference in side effects or infec-
tion rates was observed between groups. The ongoing
RITAZAREM trial analyzes another RTX maintenance
regimen (1 g every 4 months for 2 years) with conven-
tional maintenance therapy with AZA in patients with
relapsing AAV who received RTX for induction. The first
report of the trial showed a high level of efficacy (90%)
with RTX in conjunction with glucocorticoids for the re-
induction of remission in patients with AAV who have
relapsed [95]. An approach to individualized mainte-
nance therapy employing RTX dosing tailored to B-cell
reappearance and/or increase in ANCA titers was evalu-
ated in the MAINRITSAN 2 trial [96]. In this study, the
median number of infusions was reduced (3 vs. 5) in the
tailored-dosegroupcomparedtopatientsreceivingfixed-
dose RTX every 6 months. Although not significant, a
trend toward a higher rate of relapse in patients with
RTX-tailored dosing (17.3%) compared with patients re-
ceiving fixed-dose RTX (9.9%) was observed. Interesting-
ly, 50% of the relapses in this study occurred in patients
without B-lymphocyte reconstitution and 30% with neg-
ative ANCA, highlighting the limitation of these bio-
markers to predict relapse. Notably, up to 30% of patients
experienced a relapse 38 months after the last RTX infu-
sion, putting into debate the optimal duration of mainte-
nance immunosuppression.
In this sense, the REMAIN trial was a randomized con-
trolled trial that tested whether continuing AZA and
prednisone treatment for 48 months was more effective
in relapse prevention than withdrawal at 24 months [97].
Results from this study showed a significant decrease in
both major and minor relapses and better renal survival
in the continuation group. More recently, the MAINRIT-
SAN 3 trial included PR3 and MPO-AAV patients who
were in complete remission after receiving an 18-month
RTX maintenance regimen in the MAINRITSAN 2 study
[98], and participants were randomly assigned to receive
prolonged maintenance therapy with either intravenous
RTX (500 mg every 6 months) or a placebo for a further
18 months. Relapse-free survival rates at month 28 were
significantly higher among patients given prolonged RTX
(96%) than patients given the placebo (74%), and long-
term RTX maintenance therapy did not increase the
number of adverse events. Notably, relapses occurred
more frequently in PR3-ANCA patients than in MPO pa-
tients in the placebo arm (40% vs. 12%). On the basis of
these results, prolonged maintenance treatment (4–5
years) with RTX may be employed at high-risk patients
for relapses, such as those with PR3-ANCA, those who
have already had a relapse, or with persistent hematuria
or high ANCA titers despite treatment.
Refractory Disease and Other Therapies
Refractory disease in AAV can be defined as progres-
sive disease within the first 3 months or lack control of
vasculitis activity by 6 months despite standard of care
therapy. An increase in the glucocorticoid dose is used in
severe disease, but prolonged use of high-dose oral gluco-
corticoids should be avoided due to the associated risks.
Switching from CYC to RTX and vice versa or a combina-
tion of both immunosuppressants could be considered in
refractory cases.
The development of anti-RTX antibodies has been de-
scribed to neutralize RTX B-cell cytotoxicity and may im-
pact the clinical outcome of autoimmune diseases [99].
Ofatumumab, a fully humanized antibody directed
against a distinct extracellular epitope of CD20 has slow-
er dissociation kinetics than that of RTX and has been
shown to be a more potent activator of complement-de-
pendent cytotoxicity in vitro. It was successfully em-
ployed in small series of AAV cases [100], but its role in
refractory AAV disease requires further investigation.
ANCA Vasculitis: An Individualized
Approach
133
Nephron 2022;146:121–137
DOI: 10.1159/000520727
The efficacy and safety of mepolizumab, a human
monoclonal antibody against IL-5, for the treatment of
relapsing or refractory EGPA, were investigated in a ran-
domized, placebo-controlled study which included pa-
tients on a stable corticosteroid therapy [101]. Patients
treated with mepolizumab achieved a significantly higher
rate of remission than patients who received the placebo.
Also beneficial effects of IL-6 receptor blockade therapy
with tocilizumab in association with steroids have been
observed in small series of MPA patients [102], but larger
and prospective studies are desirable to confirm these re-
sults.
Given the considerable evidence for the role of TNF-α
in pathogenesis of AAV, several anti-TNF-α agents have
been explored for AAV therapy. The WGET study was a
randomized, placebo-controlled study which examined
the use of etanercept as add-on therapy to conventional
treatment, for maintenance of remission, in a large cohort
of patients with GPA [103]. This study found no addi-
tional benefit of etanercept, and excess of solid cancers
was observed, but this risk could not be attributed solely
to etanercept treatment since previous history of malig-
nancy and CYC exposure previous to etanercept could
have also played a role. Another anti-TNF agent, inflix-
imab, was evaluated as adjuvant therapy with CYC and
corticosteroids in a prospective clinical trial [104]. Inflix-
imab was effective at inducing remission in 88% of pa-
tients and permitted reduction in steroid doses. However,
severe infections were seen in 21% of patients, and despite
continued infliximab, 20% of initial responders experi-
enceddiseaseflares.Adalimumabwassimilarlyemployed
as adjuvant therapy showing high rates of early remission
(80% at 3 months) and the ability to significantly reduce
the steroid dose during induction treatment [105].
Other therapeutic approaches include the combina-
tion of the BLyS/BAFF blockade with RTX, which is now
being evaluated in the “Rituximab and Belimumab Com-
bination Therapy in PR3 Vasculitis” trial [106]. Alemtu-
zumab, a humanized anti-CD52 (CAMPATH-1H)
monoclonal antibody, was evaluated in combination with
prednisone for patients with AAV refractory to conven-
tional treatment [107]. In this study, 85% of patients
achieved remission, but 71% of these had relapsed by 9.2
months and adverse events were common.
Leflunomide was reported efficacious for remission
induction among 62 out of 93 patients (67%) with differ-
ent vasculitic disorders (64% with GCA, 89% with TAK,
80% with PAN, 69% with GPA, 75% with MPA, and 33%
with EGPA) [108]. In this study, 20% of patients discon-
tinued leflunomide before achieving remission because of
persistent disease activity and adverse events (gastroin-
testinal symptoms being the most common) led to drug
discontinuation in 18 (19%) patients.
Antithymocyte globulin (ATG) was also employed as
adjuvant therapy in a small cohort of patients with refrac-
tory GPA [109]. The majority of patients achieved remis-
sion after therapy, and treatment with ATG allowed a fur-
ther reduction in immunosuppression. However, authors
recommended avoiding ATG therapy if simultaneous in-
fections, fluid overload, or alveolar hemorrhage were
present. Also in GPA patients with refractory disease, a
6-month course of the immunosuppressant 15-deoxys-
pergualin was successfully employed in 7 patients [110].
However, most patients relapsed after 15-deoxyspergua-
lin discontinuation.
There is little experience with agents targeting plasma
cells in AAV. Experimental studies showed that bortezo-
mib depleted MPO-specific plasma cells and prevented
anti-MPO IgG-mediated necrotizing crescentic glomeru-
lonephritis in mice [111]. Interestingly, a favorable effect
of proteasome inhibition was reported in 1 patient with
AAV [112].
Finally, the use of the Ig was analyzed in a randomized,
placebo-controlled trial that investigated the efficacy of a
single course of intravenous Ig (IVIg) (total dose 2 g/kg)
in previously treated AAV patients, showing persistent
disease activity [113]. A single course of IVIg reduced dis-
ease activity in these patients, but the effect was not main-
tained beyond 3 months. Therefore, IVIg may be an alter-
native treatment option for AAV with persistent disease
activity after standard therapy. In this sense, the ongoing
Endurance 1 trial is a randomized and prospective study
which aims to prove the superiority of combination treat-
ment RTX with CYC compared to standard of care ther-
apy in AAV patients showing persistent disease activity
[114].
Conclusions
AAVs have a complex pathogenesis, influenced by ge-
netics and environmental factors. There is evidence for a
loss of tolerance to neutrophil proteins, which leads to
ANCA-mediated neutrophil activation, recruitment, and
injury, with effector T cells and activation of alternative
pathway of complement also involved. NETs containing
MPO and PR3 contribute to the pathogenesis through
different mechanisms, including antigen presentation
that promotes autoimmunity as well as direct vascular
damage.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
134
DOI: 10.1159/000520727
Data from epidemiological outcomes and genetic
studies in AAV suggest that patients should be classified
by ANCA specificity (MPO-ANCA vs. PR3-ANCA) rath-
er than by the clinical syndrome. Novel biomarkers for
screening disease, monitoring therapeutic interventions
and identifying relapses include neutrophil activation
markers such as calprotectin, macrophage activation-de-
rived products (uCD163 and MCP), T-cell subpopula-
tions, and alternative pathway of complement fractions.
The treatment of AAV is rapidly evolving, and mortal-
ity and remission rates have improved over the years.
However, high infection rates are still observed, and cur-
rent strategies are focused on reducing treatment toxicity.
RTX-based regimens are the cornerstone for induction
therapy among PR3 and relapsing patients as well as for
maintenance therapy. Novel therapeutic strategies in-
clude blockage of complement and combination of im-
munosuppressants, which allow minimization of cumu-
lative steroids exposure. Individualization of treatment
according to ANCA specificity, disease phenotype, and
risk of relapse is a key for offering a personalized care to
AAV patients.
Acknowledgments
We thank all the members of the Spanish Group for the Study
of Glomerular Diseases (GLOSEN) for their valuable contribution
to research in glomerular diseases.
Conflict of Interest Statement
The authors have no conflicts of interest to declare.
Funding Sources
There were no funding sources for this work.
Author Contributions
All the authors contributed equally to this study, revised the
paper, and approved the final version of the manuscript.
References
  1	 Rowaiye OO, Kusztal M, Klinger M. The kid-
neys and ANCA-associated vasculitis: from
pathogenesis to diagnosis. Clin Kidney J. 2015
Jun;8(3):343–50.
  2	 Jennette JC, Falk RJ, Bacon PA, Basu N, Cid
MC, Ferrario F, et al. 2012 revised Interna-
tional Chapel Hill Consensus Conference No-
menclature of Vasculitides. Arthritis Rheum.
2013 Jan;65(1):1–11.
  3	 Jennette JC, Nachman PH. ANCA glomeru-
lonephritis and vasculitis. Clin J Am Soc
Nephrol. 2017 Oct;12(10):1680–91.
 4	 Lyons PA, Rayner TF, Trivedi S, Holle JU,
Watts RA, Jayne DRW, et al. Genetically dis-
tinct subsets within ANCA-associated vascu-
litis. N Engl J Med. 2012 Jul;367(3):214–23.
  5	 Yates M, Watts R. ANCA-associated vasculi-
tis. Clin Med. 2017 Feb;17(1):60.
  6	 Berti A, Dejaco C. Update on the epidemiol-
ogy, risk factors, and outcomes of systemic
vasculitides. Best Pract Res Clin Rheumatol.
2018;32(2):271–94.
  7	 Watts RA, Robson J. Introduction, epidemi-
ology and classification of vasculitis. Best
Pract Res Clin Rheumatol. 2018;32(1):3–20.
  8	 Fujimoto S, Watts RA, Kobayashi S, Suzuki K,
Jayne DRW, Scott DGI, et al. Comparison of
the epidemiology of anti-neutrophil cytoplas-
mic antibody-associated vasculitis between
Japan and the U.K. Rheumatology. 2011 Oct;
50(10):1916–20.
  9	 Popa ER, Stegeman CA, Abdulahad WH, van
der Meer B, Arends J, Manson WM, et al.
Staphylococcal toxic-shock-syndrome-tox-
in-1 as a risk factor for disease relapse in We-
gener’s granulomatosis. Rheumatology. 2007
Jun;46(6):1029–33.
10	 Gómez-Puerta JA, Gedmintas L, Costenbader
KH. The association between silica exposure
and development of ANCA-associated vascu-
litis: systematic review and meta-analysis.
Autoimmun Rev. 2013 Oct;12(12):1129–35.
11	 Clarkson AR, Robson JS. The effect of drugs
on immunological renal disease. Prog Bio-
chem Pharmacol. 1972;7:427–51.
12	 Draibe J, Rodó X, Fulladosa X, Martínez-Va-
lenzuelaL,Diaz-EncarnaciónM,SantosL,etal.
Seasonal variations in the onset of positive and
negative renal ANCA-associated vasculitis in
Spain. Clin Kidney J. 2018 Aug;11(4):468–73.
13	 Rahmattulla C, Mooyaart AL, van Hooven D,
Schoones JW, Bruijn JA, Dekkers OM, et al.
Genetic variants in ANCA-associated vascu-
litis: a meta-analysis. Ann Rheum Dis. 2016
Sep;75(9):1687–92.
14	 Mayadas TN, Cullere X, Lowell CA. The mul-
tifaceted functions of neutrophils. Annu Rev
Pathol. 2014;9:181–218.
15	 Wilde B, Thewissen M, Damoiseaux J, van
Paassen P, Witzke O, Tervaert JW. T cells in
ANCA-associated vasculitis: what can we
learn from lesional versus circulating T cells?
Arthritis Res Ther. 2010;12(1):204.
16	Kessenbrock K, Krumbholz M, Schöner-
marck U, Back W, Gross WL, Werb Z, et al.
Netting neutrophils in autoimmune small-
vessel vasculitis. Nat Med. 2009 Jun;15(6):
623–5.
17	 Aratani Y. Myeloperoxidase: its role for host
defense, inflammation, and neutrophil func-
tion. Arch Biochem Biophys. 2018 Feb;640:
47–52.
18	 Kain R, Exner M, Brandes R, Ziebermayr R,
Cunningham D, Alderson CA, et al. Molecu-
lar mimicry in pauci-immune focal necrotiz-
ing glomerulonephritis. Nat Med. 2008 Oct;
14(10):1088–96.
19	 Pendergraft WF, Preston GA, Shah RR, Trop-
sha A, Carter CW, Jennette JC, et al. Autoim-
munity is triggered by cPR-3(105-201), a pro-
tein complementary to human autoantigen
proteinase-3. Nat Med. 2004 Jan;10(1):72–9.
20	 Balding CE, Howie AJ, Drake-Lee AB, Savage
CO. Th2 dominance in nasal mucosa in pa-
tients with Wegener’s granulomatosis. Clin
Exp Immunol. 2001 Aug;125(2):332–9.
21	 Ruth AJ, Kitching AR, Kwan RYQ, Odobasic
D, Ooi JDK, Timoshanko JR, et al. Anti-neu-
trophil cytoplasmic antibodies and effector
CD4+ cells play nonredundant roles in anti-
myeloperoxidase crescentic glomerulone-
phritis. J Am Soc Nephrol. 2006 Jun;17(7):
1940–9.
22	 Walsh M, Chaudhry A, Jayne D. Long-term
follow-up of relapsing/refractory anti-neu-
trophil cytoplasm antibody associated vascu-
litis treated with the lymphocyte depleting an-
tibody alemtuzumab (CAMPATH-1H). Ann
Rheum Dis. 2007 Nov;67(9):1322–7.
ANCA Vasculitis: An Individualized
Approach
135
Nephron 2022;146:121–137
DOI: 10.1159/000520727
23	 Abdulahad WH, van der Geld YM, Stegeman
CA, Kallenberg CG. Persistent expansion of
CD4+ effector memory T cells in Wegener’s
granulomatosis. Kidney Int. 2006 Sep;70(5):
938–47.
24	 Abdulahad WH, Stegeman CA, Limburg PC,
Kallenberg CG. Skewed distribution of Th17
lymphocytes in patients with Wegener’s gran-
ulomatosis in remission. Arthritis Rheum.
2008 Jul;58(7):2196–205.
25	 Reijnders TDY, Stegeman CA, Huitema MG,
Rutgers A, Heeringa P, Abdulahad WH. Un-
raveling the identity of FoxP3+ regulatory T
cells in granulomatosis with polyangiitis pa-
tients. Sci Rep. 2019;9(1):8273–9.
26	 McKinney EF, Lyons PA, Carr EJ, Hollis JL,
Jayne DRW, Willcocks LC, et al. A CD8+ T
cell transcription signature predicts progno-
sis in autoimmune disease. Nat Med. 2010
May;16(5):586–91.
27	 Moosig F, Csernok E, Wang G, Gross WL.
Costimulatory molecules in Wegener’s gran-
ulomatosis (WG): lack of expression of CD28
and preferential up-regulation of its ligands
B7-1 (CD80) and B7-2 (CD86) on T cells. Clin
Exp Immunol. 1998 Oct;114(1):113–8.
28	 Moiseev S, Lee JM, Zykova A, Bulanov N,
Novikov P, Gitel E, et al. The alternative com-
plement pathway in ANCA-associated vascu-
litis: further evidence and a meta-analysis.
Clin Exp Immunol. 2020 Dec;202(3):394–
402.
29	Geetha D, Jefferson JA. ANCA-associated
vasculitis: core curriculum 2020. Am J Kidney
Dis. 2020 Jan;75(1):124–37.
30	 Jayne DRW, Merkel PA, Schall TJ, Bekker P.
Avacopan for the treatment of ANCA-associ-
ated vasculitis. N Engl J Med. 2021 Feb;
384(7):599–609.
31	 Said MS. Upper respiratory tract symptoms,
renal involvement and vasculitis: a case report
and review of wegener granulomatosis. J Clin
Med Res. 2010 Aug;2(4):189–93.
32	Parra-García GD, Callejas-Rubio JL, Ríos-
Fernández R, Sainz-Quevedo M, Ortego-
Centeno N. Otolaryngologic manifestations
of systemic vasculitis. Acta Otorrinolaringol.
2012 Jul;63(4):303–10.
33	 Watkins AS, Kempen JH, Choi D, Liesegang
TL, Pujari SS, Newcomb C, et al. Ocular dis-
ease in patients with ANCA-positive vasculi-
tis. J Ocul Biol Dis Inform. 2009 Dec;3(1):
12–9.
34	 Demirkesen C. Approach to cutaneous vascu-
litides with special emphasis on small vessel
vasculitis: histopathology and direct immu-
nofluorescence. Curr Op Rheumatol. 2017
Jan;29(1):39–44.
35	Wludarczyk A, Szczeklik W. Neurological
manifestations in ANCA-associated vasculi-
tis: assessment and treatment. Expert Rev
Neurother. 2016 Aug;16(8):861–3.
36	 Storesund B, Gran JT, Koldingsnes W. Severe
intestinal involvement in Wegener’s granulo-
matosis: report of two cases and review of the
literature. Br J Rheumatol. 1998 Apr;37(4):
387–90.
37	 Hunder GG, Arend WP, Bloch DA, Calabrese
LH, Fauci AS, Fries JF, et al. The American
College of Rheumatology 1990 criteria for the
classification of vasculitis: introduction. Ar-
thritis Rheum. 1990;33(8):1065–7.
38	 Rasmussen N, Wiik A. Indirect immunofluo-
rescence examination for IgG-ANCA in sera
submitted for the 1st international workshop
on ANCA, 1988. APMIS Suppl. 1989;6:16–
20.
39	Schulte-Pelkum J, Radice A, Norman GL,
Lόpez Hoyos M, Lakos G, Buchner C, et al.
Novel clinical and diagnostic aspects of anti-
neutrophil cytoplasmic antibodies. J Immu-
nol Res. 2014;2014:185416.
40	 Savige J, Gillis D, Benson E, Davies D, Esnault
V, Falk RJ, et al. International consensus
statement on testing and reporting of anti-
neutrophil cytoplasmic antibodies (ANCA).
Am J Clin Pathol. 1999 Apr;111(4):507–13.
41	 Chung SA, Seo P. Microscopic polyangiitis.
Rheum Dis Clin North Am. 2010 Aug;36(3):
545–58.
42	 Lutalo PMK, D’Cruz DP. Diagnosis and clas-
sification of granulomatosis with polyangiitis
(aka Wegener’s granulomatosis). J Autoim-
mun. 2014 Feb;48:94–8.
43	 Gioffredi A, Maritati F, Oliva E, Buzio C. Eo-
sinophilic granulomatosis with polyangiitis:
an overview. Front Immunol. 2014;5:549.
44	 Yates M, Watts R, Bajema I, Cid M, Crestani
B, Hauser T, et al. Validation of the EULAR/
ERA-EDTA recommendations for the man-
agement of ANCA-associated vasculitis by
disease content experts. RMD open. 2017 Jun;
3(1):e000449.
45	 Mukhtyar C, Lee R, Brown D, Carruthers D,
Dasgupta B, Dubey S, et al. Modification and
validation of the birmingham vasculitis activ-
ity score (version 3). Ann Rheum Dis. 2009
Dec;68(12):1827–32.
46	 Guillevin L, Pagnoux C, Seror R, Mahr A,
Mouthon L, Toumelin PL. The five-factor
score revisited: assessment of prognoses of
systemic necrotizing vasculitides based on the
french vasculitis study group (FVSG) cohort.
Medicine. 2011 Jan;90(1):19–27.
47	 Exley AR, Bacon PA, Luqmani RA, Kitas GD,
Gordon C, Savage COS, et al. Development
and initial validation of the vasculitis damage
index for the standardized clinical assessment
of damage in the systemic vasculitides. Ar-
thritis Rheumat. 1997;40(2):371–80.
48	 Kemna MJ, Damoiseaux J, Austen J, Winkens
B, Peters J, van Paassen P, et al. ANCA as a
predictor of relapse: useful in patients with re-
nal involvement but not in patients with non-
renal disease. J Am Soc Nephrol. 2015 Mar;
26(3):537–42.
49	 Lionaki S, Blyth ER, Hogan SL, Hu Y, Senior
BA, Jennette CE, et al. Classification of anti-
neutrophil cytoplasmic autoantibody vasculi-
tides: the role of antineutrophil cytoplasmic
autoantibody specificity for myeloperoxidase
or proteinase 3 in disease recognition and
prognosis. Arthritis Rheumat. 2012 Oct;
64(10):3452–62.
50	 Hong Y, Eleftheriou D, Hussain AAK, Price-
Kuehne FE, Savage CO, Jayne D, et al. Anti-
neutrophil cytoplasmic antibodies stimulate
release of neutrophil microparticles. J Am Soc
Nephrol. 2012;23:49–62.
51	 Wang H, Sha LL, Ma TT, Zhang LX, Chen M,
Zhao MH. Circulating level of neutrophil ex-
tracellular traps is not a useful biomarker for
assessing disease activity in antineutrophil cy-
toplasmic antibody-associated vasculitis.
PLoS One. 2016 Feb 3;11(2):e0148197.
52	 Pepper RJ, Wang HH, Rajakaruna GK, Pa-
pakrivopoulou E, Vogl T, Pusey CD, et al.
S100A8/A9 (calprotectin) is critical for devel-
opment of glomerulonephritis and promotes
inflammatory leukocyte-renal cell interac-
tions. Am J Pathol. 2015 May;185(5):1264–
74.
53	 Pepper RJ, Hamour S, Chavele KM, Todd SK,
Rasmussen N, Flint S, et al. Leukocyte and se-
rum S100A8/S100A9 expression reflects dis-
ease activity in ANCA-associated vasculitis
and glomerulonephritis. Kidney Int. 2013
Jun;83(6):1150–8.
54	 Martinez Valenzuela L, Draibe J, Ramos MQ,
Fulladosa Oliverasid X, Melilli E, Cruzado
Garrit JM, et al. Calprotectin as a smoldering
activity detection tool and renal prognosis
biomarker in ANCA associated vasculitis.
PLoS One. 2018;13(10).
55	 O’Reilly VP, Wong L, Kennedy C, Elliot LA,
O’Meachair S, Coughlan AM, et al. Urinary
soluble CD163 in active renal vasculitis. J Am
Soc Nephrol. 2016 Sep;27(9):2906–16.
56	 Villacorta J, Lucientes L, Goicoechea E, Ace-
vedo M, Cavero T, Sanchez-Camara L, et al.
Urinary soluble CD163 as a biomarker of
disease activity and relapse in antineutro-
phil cytoplasm antibody-associated glomer-
ulonephritis. Clin Kidney J. 2021 Feb;14(1):
212–9.
57	 Dekkema GJ, Abdulahad WH, Bijma T, Mo-
ran SM, Ryan L, Little MA, et al. Urinary and
serum soluble CD25 complements urinary
soluble CD163 to detect active renal anti-neu-
trophil cytoplasmic autoantibody-associated
vasculitis: a cohort study. Nephrol Dial
Transplant. 2019 Feb;34(2):234–24.
58	 Moran SM, Monach PA, Zgaga L, Cuthbert-
son D, Carette S, Khalidi NA, et al. Urinary
soluble CD163 and monocyte chemoattrac-
tant protein-1 in the identification of subtle
renal flare in anti-neutrophil cytoplasmic an-
tibody–associated vasculitis. Nephrol Dial
Transplant. 2020 Feb;35(2):283–91.
59	 Valenzuela LM, Draibe J, Quero M, Fulladosa
X, Cruzado JM, Bestard O, et al. Exploring
frequencies of circulating specific th17 cells
against myeloperoxidase and proteinase 3 in
anca associated vasculitis. Int J Mol Sci. 2019
Dec;20(23):5820.
60	Morgan MD, Day CJ, Piper KP, Khan N,
Harper L, Moss PA, et al. Patients with We-
gener’s granulomatosis demonstrate a rela-
tive deficiency and functional impairment of
T-regulatory cells. Immunology. 2010 May;
130(1):64–73.
Villacorta/Martinez-Valenzuela/
Martin-Capon/Bordignon-Draibe
Nephron 2022;146:121–137
136
DOI: 10.1159/000520727
61	 Yoshimura J, Fukami K, Koike K, Nagano M,
Matsumoto T, Iwatani R, et al. Interstitial
Foxp3-positive T cells may predict renal sur-
vival in patients with myeroperoxidase anti-
neutrophil cytoplasmic antibody-associated
glomerulonephritis. Clin Exp Pharmacol
Physiol. 2010 Sep;37(9):879–83.
62	Villacorta J, Diaz-Crespo F, Acevedo M,
Cavero T, Guerrero C, Praga M, et al. Circu-
lating C3 levels predict renal and global out-
come in patients with renal vasculitis. Clin
Rheumatol. 2016 Nov;35(11):2733–40.
63	Villacorta J, Diaz-Crespo F, Acevedo M,
Guerrero C, Campos-Martin Y, García-Díaz
E, et al. Glomerular C3d as a novel prognostic
marker for renal vasculitis. Human Pathol.
2016 Oct;56:31–9.
64	 Robson J, Doll H, Suppiah R, Flossmann O,
Harper L, Höglund P, et al. Damage in the an-
ca-associated vasculitides: long-term data
from the European vasculitis study group
(EUVAS) therapeutic trials. Ann Rheum Dis.
2015 Jan;74(1):177–84.
65	 Flossmann O, Berden A, de Groot K, Hagen
C, Harper L, Heijl C, et al. Long-term patient
survival in ANCA-associated vasculitis. Ann
Rheum Dis. 2011 Mar;70(3):488–94.
66	 de Groot K, Harper L, Jayne DRW, Flores Su-
arez LF, Gregorini G, Gross WL, et al. Pulse
versus daily oral cyclophosphamide for in-
duction of remission in antineutrophil cyto-
plasmic antibody-associated vasculitis: a ran-
domized trial. Ann Intern Med. 2009 May;
150(10):670–80.
67	 Harper L, Morgan MD, Walsh M, Hoglund P,
Westman K, Flossmann O, et al. Pulse versus
daily oral cyclophosphamide for induction of
remission in ANCA-associated vasculitis:
long-term follow-up. Ann Rheum Dis. 2012
Jun;71(6):955–60.
68	 Wester Trejo MAC, Bajema IM, van Daalen
EE. Antineutrophil cytoplasmic antibody-as-
sociated vasculitis and malignancy. Curr
Opin Rheumatol. 2018 Jan;30(1):44–9.
69	 Lafarge A, Joseph A, Pagnoux C, Puechal X,
Cohen P, Samson M, et al. Risk of malignancy
in patients treated for systemic necrotising
vasculitis. Ann Rheum Dis. 2020 Mar;79(3):
431–3.
70	 Knight A, Askling J, Granath F, Sparen P, Ek-
bom A. Urinary bladder cancer in Wegener’s
granulomatosis: risks and relation to cyclo-
phosphamide. Ann Rheum Dis. 2004 Oct;
63(10):1307–11.
71	 Emadi A, Jones RJ, Brodsky RA. Cyclophos-
phamide and cancer: golden anniversary. Nat
Rev Clin Oncol. 2009 Nov;6(11):638–47.
72	 Jones RB, Tervaert JWC, Hauser T, Luqmani
R, Morgan MD, Peh CA, et al. Rituximab ver-
sus cyclophosphamide in ANCA-associated
renal vasculitis. N Engl J Med. 2010;363(3):
211–20.
73	 Stone JH, Merkel PA, Spiera R, Seo P, Lang-
ford CA, Hoffman GS, et al. Rituximab versus
cyclophosphamide for ANCA-associated
vasculitis. N Engl J Med. 2010 Jul;363(3):
221–32.
74	 Unizony S, Villarreal M, Miloslavsky EM, Lu
N, Merkel PA, Spiera R, et al. Clinical out-
comes of treatment of anti-neutrophil cyto-
plasmic antibody (ANCA)-associated vascu-
litis based on ANCA type. Ann Rheum Dis.
2016 Jun;75(6):1166–9.
75	Yates M, Watts RA, Bajema IM, Cid MC,
Crestani B, Hauser T, et al. EULAR/ERA-ED-
TA recommendations for the management of
ANCA-associatedvasculitis.AnnRheumDis.
2016 Sep;75(9):1583–94.
76	 Rovin BH, Caster DJ, Cattran DC, Gibson KL,
Hogan JJ, Moeller MJ, et al. Management and
treatment of glomerular diseases (part 2):
conclusions from a Kidney Disease: Improv-
ing Global Outcomes (KDIGO) Controver-
sies Conference. Kidney Int. 2019 Feb;95(2):
281–95.
77	 McClure M, Gopaluni S, Jayne D, Jones R. B
cell therapy in ANCA-associated vasculitis:
current and emerging treatment options. Nat
Rev Rheumatol. 2018 Oct;14(10):580–91.
78	 Roberts DM, Jones RB, Smith RM, Alberici F,
Kumaratne DS, Burns S, et al. Rituximab-as-
sociated hypogammaglobulinemia: inci-
dence, predictors and outcomes in patients
with multi-system autoimmune disease. J Au-
toimmun. 2015 Feb;57:60–5.
79	 Tieu J, Smith R, Basu N, Brogan P, D’Cruz D,
Dhaun N, et al. Rituximab for maintenance of
remission in ANCA-associated vasculitis: ex-
pert consensus guidelines. Rheumatology.
2020 Apr;59(4):e24–32.
80	 Tsutsumi Y, Yamamoto Y, Ito S, Ohigashi H,
Shiratori S, Naruse H, et al. Hepatitis B virus
reactivation with a rituximab-containing reg-
imen. World J Hepatol. 2015 Sep;7(21):2344–
51.
81	 Loomba R, Liang TJ. Hepatitis B reactivation
associated with immune suppressive and bio-
logical modifier therapies: current concepts,
management strategies, and future directions.
Gastroenterology. 2017 May;152(6):1297–309.
82	 Zonozi R, Wallace ZS, Laliberte K, Huizenga
NR, Rosenthal JM, Rhee EP, et al. Incidence,
clinical features, and outcomes of late-onset
neutropenia from rituximab for autoimmune
disease. Arthritis Rheumatol. 2021 Feb;73(2):
347–54.
83	 Jones RB, Hiemstra TF, Ballarin J, Blockmans
DE, Brogan P, Bruchfeld A, et al. Mycopheno-
late mofetil versus cyclophosphamide for re-
mission induction in ANCA-associated vas-
culitis: a randomised, non-inferiority trial.
Ann Rheum Dis. 2019 Mar;78(3):399–405.
84	Grennan D, Wang S. Steroid side effects.
JAMA. 2019 Jul;322(3):282.
85	 McAdoo SP, Medjeral-Thomas N, Gopaluni
S, Tanna A, Mansfield N, Galliford J, et al.
Long-term follow-up of a combined ritux-
imab and cyclophosphamide regimen in renal
anti-neutrophil cytoplasm antibody-associat-
ed vasculitis. Nephrol Dial Transplant. 2019
Jan;34(1):63–73.
86	 Pepper RJ, McAdoo SP, Moran SM, Kelly D,
Scott J, Hamour S, et al. A novel glucocorti-
coid-free maintenance regimen for anti-neu-
trophil cytoplasm antibody-associated vascu-
litis. Rheumatology. 2019 Feb;58(2):373.
87	 Walsh M, Merkel PA, Peh CA, Szpirt WM,
Puéchal X, Fujimoto S, et al. Plasma exchange
and glucocorticoids in severe ANCA-associ-
ated vasculitis. N Engl J Med. 2020 Feb;
382(7):622–31.
88	 U.S National Library of Medicine. Safety and
Efficacy Study of IFX-1 in add-on to standard
of care in granulomatosis with polyangiitis
(GPA) and microscopic polyangiitis (MPA).
2021. Available from: https://clinicaltrials.
gov/ct2/show/NCT03712345.
89	 Lu J, Zhang L, Xia C, Tao Y. Complications of
therapeutic plasma exchange: a retrospective
study of 1201 procedures in 435 children.
Medicine. 2019 Dec;98(50):e18308.
90	
Jayne DRW, Gaskin G, Rasmussen N,
Abramowicz D, Ferrario F, Guillevin L, et al.
Randomized trial of plasma exchange or
high-dosage methylprednisolone as adjunc-
tive therapy for severe renal vasculitis. J Am
Soc Nephrol. 2007 Jul;18(7):2180–8.
91	 Walsh M, Casian A, Flossmann O, Westman
K, Höglund P, Pusey C, et al. Long-term fol-
low-up of patients with severe ANCA-associ-
ated vasculitis comparing plasma exchange to
intravenous methylprednisolone treatment is
unclear. Kidney Int. 2013 Aug;84(2):397–402.
92	 Salama AD. Relapse in anti-neutrophil cyto-
plasm antibody (ANCA)–associated vasculi-
tis. Kidney Int Rep. 2019 Oct 24;5(1):7–12.
93	 Jayne D, Rasmussen N, Andrassy K, Bacon P,
Tervaert JWC, Dadoniené J, et al. A random-
ized trial of maintenance therapy for vasculi-
tis associated with antineutrophil cytoplasmic
autoantibodies. N Engl J Med. 2003 Jul;
349(1):36–44.
94	 Guillevin L, Pagnoux C, Karras A, Khouatra
C, Aumaître O, Cohen P, et al. Rituximab ver-
sus azathioprine for maintenance in ANCA-
associated vasculitis. N Engl J Med. 2014 Nov;
371(19):1771–80.
95	 Smith RM, Jones RB, Specks U, Bond S, No-
dale M, Aljayyousi R, et al. Rituximab as ther-
apy to induce remission after relapse in AN-
CA-associated vasculitis. Ann Rheum Dis.
2020 Sep;79(9):1243–9.
96	 Charles P, Terrier B, Perrodeau É, Cohen P,
Faguer S, Huart A, et al. Comparison of indi-
vidually tailored versus fixed-schedule ritux-
imab regimen to maintain ANCA-associated
vasculitis remission: results of a multicentre,
randomised controlled, phase III trial
(MAINRITSAN2). Ann Rheum Dis. 2018
Aug;77(8):1143–9.
97	 Karras A, Pagnoux C, Haubitz M, Groot K,
Puechal X, Tervaert JWC, et al. Randomised
controlled trial of prolonged treatment in the
remission phase of ANCA-associated vasculi-
tis. Ann Rheum Dis. 2017 Oct;76(10):1662–8.
98	 Charles P, Perrodeau É, Samson M, Bonnotte
B, Néel A, Agard C, et al. Long-term ritux-
imab use to maintain remission of antineu-
trophil cytoplasmic antibody-associated vas-
culitis: a randomized trial. Ann Intern Med.
2020 Aug;173(3):179–87.
ANCA Vasculitis: An Individualized
Approach
137
Nephron 2022;146:121–137
DOI: 10.1159/000520727
 99	Boyer-Suavet S, Andreani M, Lateb M,
Savenkoff B, Brglez V, Benzaken S, et al.
Neutralizing anti-rituximab antibodies and
relapseinmembranousnephropathytreated
with rituximab. Front Immunol. 2019;10:
3069.
100	 McAdoo SP, Bedi R, Tarzi R, Griffith M,
Pusey CD, Cairns TD. Ofatumumab for B
cell depletion therapy in ANCA-associated
vasculitis: a single-centre case series. Rheu-
matology. 2016 Aug;55(8):1437–42.
101	 Wechsler ME, Akuthota P, Jayne D, Khoury
P,KlionA,LangfordCA,etal.Mepolizumab
or placebo for eosinophilic granulomatosis
with polyangiitis. N Engl J Med. 2017;
376(20):1921–32.
102	 Sakai R, Kondo T, Kurasawa T, Nishi E,
Okuyama A, Chino K, et al. Current clinical
evidence of tocilizumab for the treatment of
ANCA-associated vasculitis: a prospective
case series for microscopic polyangiitis in a
combination with corticosteroids and litera-
ture review. Clin Rheumatol. 2017 Oct;
36(10):2383–92.
103	 The Wegener’s Granulomatosis Etanercept
Trial (WGET) Research Group. Etanercept
in addition to standard therapy in patients
with Wegener’s granulomatosis. N Engl J
Med. 2005;352:351–61.
104	 Booth A, Harper L, Hammad T, Bacon P,
Griffith M, Levy J, et al. Prospective study of
TNF-ablockadewithinfliximabinanti-neu-
trophil cytoplasmic antibody-associated
systemic vasculitis. J Am Soc Nephrol. 2004;
15:717–72.
105	 Laurino S, Chaudhry A, Booth A, Conte G,
Jayne D. Prospective study of TNFalpha
blockade with adalimumab in ANCA-asso-
ciated systemic vasculitis with renal involve-
ment. Nephrol Dial Transplant. 2010 Oct;
25(10):3307–14.
106	 U.S National Library of Medicine. Ritux-
imab and belimumab combination therapy
in PR3 vasculitis (COMBIVAS). 2020.
Available from: https://clinicaltrials.gov/
ct2/show/NCT03967925.
107	Walsh M, Chaudhry A, Jayne D. Long-
term follow-up of relapsing/refractory an-
tineutrophil cytoplasm antibody associat-
ed vasculitis treated with the lymphocyte
depleting antibody alemtuzumab (CAM-
PATH-1H). Ann Rheum Dis. 2008;67:
1322–7.
108	 Mustapha N, Barra L, Carette S, Cuthbert-
son D, Khalidi NA, Koening CL, et al. Cana-
dian vasculitis research network (CanVasc)
and vasculitis clinical research consortium
(VCRC). Efficacy of leflunomide in the
treatment of vasculitis. Clin Exp Rheumatol.
2021;129(2):114–8.
109	Schmitt WH, Hagen EC, Neumann I,
Nowack R, Flores-Suárez LF, Van der
WoudeFJ,etal.TreatmentofrefractoryWe-
gener’s granulomatosis with antithymocyte
globulin (ATG): an open study in 15 pa-
tients. Kidney Int. 2004;65:1440–8.
110	 Flossmann O, Jayne DR. Long-term treat-
ment of relapsing Wegener’s granulomato-
sis with 15-deoxyspergualin. Rheumatology.
2010 Mar;49(3):556–62.
111	 Bontscho J, Schreiber A, Manz RA, Schnei-
der W, Luft FC, Kettritz R. Myeloperoxi-
dase-specific plasma cell depletion by
bortezomib protects from anti-neutrophil
cytoplasmic autoantibodies-induced glo-
merulonephritis. J Am Soc Nephrol. 2011
Feb;22(2):336–48.
112	 Novikov P, Moiseev S, Bulanov N, Shchegol-
eva E. Bortezomib in refractory ANCA-as-
sociated vasculitis: a new option? Ann
Rheum Dis. 2016 Jan;75(1):e9.
113	Jayne DR, Chapel H, Adu D, Misbah S,
O’Donoghue D, Scott D, et al. Intravenous
immunoglobulin for ANCA-associated sys-
temic vasculitis with persistent disease activ-
ity. QJM. 2000 Jul;93(7):433–9.
114	 U.S National Library of Medicine. Exploring
durable remission with rituximab in anti-
neutrophil cytoplasmic antibody(ANCA)-
associated vasculitis (ENDURRANCE-1).
2021. Available from: https://clinicaltrials.
gov/ct2/show/NCT03942887?term=
ENDURRANCE&draw=2&rank=1.

More Related Content

PDF
ANCA vasculitis (KDIGO 2021 Guidelines) - Dr. Gawad
PPTX
Approach to Hematuria
PPTX
Metabolic Nephropathy - Dr. Gawad
PPTX
Hepatitis C virus & chronic kidney diseases
PDF
Dysproteinemias Related Renal Disorders, Monoclonal Gammopathy (Paraproteinem...
PPTX
Approach to hematuria
PPT
CMV in Organ Transplantation
PPTX
IntraDialytic Hypertension (Basic Science → Evidence → Practice) - Dr. Gawad
ANCA vasculitis (KDIGO 2021 Guidelines) - Dr. Gawad
Approach to Hematuria
Metabolic Nephropathy - Dr. Gawad
Hepatitis C virus & chronic kidney diseases
Dysproteinemias Related Renal Disorders, Monoclonal Gammopathy (Paraproteinem...
Approach to hematuria
CMV in Organ Transplantation
IntraDialytic Hypertension (Basic Science → Evidence → Practice) - Dr. Gawad

What's hot (20)

PPTX
Crystalline nephropathies
PPTX
Autoimmune Hepatitis
PPTX
Acute kidney injury
PPTX
Anca vasculitis & anti gbm
PDF
CKD Progression (Pharmacological Approach) - Dr. Gawad
PPTX
Dr hamada alsedawy sepsis and aki
PPTX
Antitubercular agents in TB patients with Chronic Liver disease (CLD)
PPTX
Cytomegalovirus infection in critically ill patients: Focus on HSCT recipients
PPTX
Presentation: HCV in dialysis
PDF
PATHOPHYSIOLOGY OF DIABETIC NEPHROPATHY
PPTX
9_Fraser_VAP_Hopkins_Fellows_Course-converted.pptx
PPTX
Fanconi anemia
PPTX
Hypernatremia - Stepwise Practical Approach - Dr. Gawad
PPTX
Chronic Hepatitis B Infection
PPT
Membranous Nephropathy
PPTX
lab work up for hemolytic anemia
PDF
Hyponatremia (Practical Approach) - Dr. Gawad
PPT
Indications of peritoneal dialysis
PPTX
Jaundice
PPTX
Syndrome of inappropriate antidiuretic hormone secretion
Crystalline nephropathies
Autoimmune Hepatitis
Acute kidney injury
Anca vasculitis & anti gbm
CKD Progression (Pharmacological Approach) - Dr. Gawad
Dr hamada alsedawy sepsis and aki
Antitubercular agents in TB patients with Chronic Liver disease (CLD)
Cytomegalovirus infection in critically ill patients: Focus on HSCT recipients
Presentation: HCV in dialysis
PATHOPHYSIOLOGY OF DIABETIC NEPHROPATHY
9_Fraser_VAP_Hopkins_Fellows_Course-converted.pptx
Fanconi anemia
Hypernatremia - Stepwise Practical Approach - Dr. Gawad
Chronic Hepatitis B Infection
Membranous Nephropathy
lab work up for hemolytic anemia
Hyponatremia (Practical Approach) - Dr. Gawad
Indications of peritoneal dialysis
Jaundice
Syndrome of inappropriate antidiuretic hormone secretion
Ad

Similar to anca vasculitis.pdf (20)

PDF
Vasculitis nrrheum.2014.103
PPTX
Anca associated vasculitis (renal and systemic)
PPTX
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
PPTX
ANCA GN.pptx
PDF
ARTICULO CLASIFICACION DE LAS VASCULITIS
PDF
ARTICULO CRITERIOS DE CLASIFICACION DE LAS VASCULITIS
PDF
CRITERIOS DE CLASIFICACION VASCULITIS SSS
PPTX
Pulmonary renal syndrome by Dr Bharat Rewaria
PPT
ANCA vasculitis
PPTX
anca vasculitis in pediaric pts .pptx
PPTX
ANCA vasculitis and Renal Diseases
PPTX
Anca talk
PDF
Vasculitis nrrheum.2014.104
PPTX
Granulomatosis polyangitis GPA
PPTX
PPTX
Vasculitis only
PPTX
Pulmonary vasculitis
PPT
Systemic vasculitis & Alpha-1 antitrypsin deficiency_ Gema Tirado-Conde
PPTX
Approach to the patient vasculitis (2)
Vasculitis nrrheum.2014.103
Anca associated vasculitis (renal and systemic)
Genetically Distinct differences of ANCA-Associated Vasculitis Disorders
ANCA GN.pptx
ARTICULO CLASIFICACION DE LAS VASCULITIS
ARTICULO CRITERIOS DE CLASIFICACION DE LAS VASCULITIS
CRITERIOS DE CLASIFICACION VASCULITIS SSS
Pulmonary renal syndrome by Dr Bharat Rewaria
ANCA vasculitis
anca vasculitis in pediaric pts .pptx
ANCA vasculitis and Renal Diseases
Anca talk
Vasculitis nrrheum.2014.104
Granulomatosis polyangitis GPA
Vasculitis only
Pulmonary vasculitis
Systemic vasculitis & Alpha-1 antitrypsin deficiency_ Gema Tirado-Conde
Approach to the patient vasculitis (2)
Ad

Recently uploaded (20)

PPTX
This book is about some common childhood
PPTX
Communicating with the FDA During an Inspection -August 26, 2025 - GMP.pptx
PDF
heliotherapy- types and advantages procedure
PDF
communicable diseases for healthcare - Part 1.pdf
PPT
fiscal planning in nursing and administration
PPTX
critical care nursing 12.pptxhhhhhhhhjhh
PPTX
A Detailed Physiology of Endocrine System.pptx
PPTX
Peripheral Arterial Diseases PAD-WPS Office.pptx
PPTX
ACUTE PANCREATITIS combined.pptx.pptx in kids
DOCX
ORGAN SYSTEM DISORDERS Zoology Class Ass
PDF
Nematodes - by Sanjan PV 20-52.pdf based on all aspects
PPTX
Approch to weakness &paralysis pateint.pptx
PDF
Diabetes mellitus - AMBOSS.pdf
PDF
Integrating Traditional Medicine with Modern Engineering Solutions (www.kiu....
PPTX
Acute Abdomen and its management updates.pptx
PPTX
Indications for Surgical Delivery...pptx
PPTX
IMMUNITY ... and basic concept mds 1st year
PPTX
GAIT IN HUMAN AMD PATHOLOGICAL GAIT ...............
PPTX
Biostatistics Lecture Notes_Dadason.pptx
PPTX
SUMMARY OF EAR, NOSE AND THROAT DISORDERS INCLUDING DEFINITION, CAUSES, CLINI...
This book is about some common childhood
Communicating with the FDA During an Inspection -August 26, 2025 - GMP.pptx
heliotherapy- types and advantages procedure
communicable diseases for healthcare - Part 1.pdf
fiscal planning in nursing and administration
critical care nursing 12.pptxhhhhhhhhjhh
A Detailed Physiology of Endocrine System.pptx
Peripheral Arterial Diseases PAD-WPS Office.pptx
ACUTE PANCREATITIS combined.pptx.pptx in kids
ORGAN SYSTEM DISORDERS Zoology Class Ass
Nematodes - by Sanjan PV 20-52.pdf based on all aspects
Approch to weakness &paralysis pateint.pptx
Diabetes mellitus - AMBOSS.pdf
Integrating Traditional Medicine with Modern Engineering Solutions (www.kiu....
Acute Abdomen and its management updates.pptx
Indications for Surgical Delivery...pptx
IMMUNITY ... and basic concept mds 1st year
GAIT IN HUMAN AMD PATHOLOGICAL GAIT ...............
Biostatistics Lecture Notes_Dadason.pptx
SUMMARY OF EAR, NOSE AND THROAT DISORDERS INCLUDING DEFINITION, CAUSES, CLINI...

anca vasculitis.pdf

  • 1. Clinical Practice: Review Article Nephron 2022;146:121–137 Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: Toward an Individualized Approach Javier Villacorta  a Laura Martinez-Valenzuela  b Irene Martin-Capon  a Juliana Bordignon-Draibe  b aNephrology Department, Ramón y Cajal University Hospital, IRYCIS, Alcala University, Madrid, Spain; b Nephrology Department, Bellvitge University Hospital, IDIBELL Biomedical Research Institute, Hospitalet de Llobregat, Barcelona, Spain Received: September 14, 2021 Accepted: October 29, 2021 Published online: December 15, 2021 Correspondence to: Javier Villacorta, javier.villacorta@salud.madrid.org © 2021 S. Karger AG, Basel karger@karger.com www.karger.com/nef DOI: 10.1159/000520727 Keywords Glomerulonephritis · Antineutrophil cytoplasmic antibody · Vasculitis Abstract Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), characterized by the presence of autoanti- bodies to neutrophil cytoplasmic antigens, proteinase 3 (PR3), and myeloperoxidase (MPO), typically involves small blood vessels of the respiratory tract and kidneys. It includes distinct clinical syndromes: microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA), and eosinophilic GPA. GPA is commonly associated with PR3-ANCA, while MPA is associated with MPO-ANCA. AAVs have a complex pathogenesis, influenced by genetics and environmental factors. There is evidence for a loss of tolerance to neutrophil proteins, which leads to ANCA-mediated neutrophil activa- tion and injury, with effector T cells and activation of the al- ternative pathway of the complement also involved. Ad- vances in immunosuppressive treatment have drastically re- duced mortality of AAV in the past decades, opting for a more individualized approach. Careful assessment of ANCA specificity, disease activity, organ damage, and quality of life allows for a tailored immunosuppressive therapy. Contem- porary AAV treatment is characterized by regimens that min- imize the cumulative exposure to glucocorticoids and cyclo- phosphamide, and novel approaches including comple- ment blockage and immunosuppressant combinations might be the standard of care in the future. In this review, we examine the pathogenesis, clinical approach, and evidence- based treatment options for the management of AAV pa- tients. © 2021 S. Karger AG, Basel Introduction Antineutrophil cytoplasmic antibody (ANCA)-associ- ated vasculitis (AAV) is a group of disorders character- ized by inflammation and destruction of small- and me- dium-sized blood vessels and the presence of circulating ANCAs [1]. It comprises distinct clinical syndromes: mi- croscopic polyangiitis (MPA), granulomatosis with poly- angiitis (GPA), and eosinophilic GPA (EGPA) [2]. Sero- logical classification of AAV into proteinase 3-ANCA (PR3-ANCA) disease and myeloperoxidase-ANCA (MPO-ANCA) disease is crucial [3]. GPA is commonly
  • 2. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 122 DOI: 10.1159/000520727 associated with PR3-ANCA, while MPA is associated with MPO-ANCA. However, differences in genetics, pathogenesis, risk factors, treatment responses, and out- comes associate closely with ANCA specificity (PR3 vs. MPO-ANCA) than with clinical syndrome [4]. There- fore, the ANCA serotype may guide clinical and thera- peutic approaches, leading to a more personalized medi- cine for AAV patients. Renal involvement is present in the majority of patients with AAV, and the consequences of a missed or delayed diagnosis of renal vasculitis are po- tentially life-threatening. Advances in immunosuppres- sive treatment have drastically reduced mortality of AAV in the past decades with higher rates of remission ob- served in recent trials [5]. However, infection rates are still high, and current regimens aim to minimize the cu- mulative exposure to glucocorticoids and cyclophospha- mide (CYC). In this review, we discuss the pathogenesis, clinical classification, diagnostic approach, potential bio- markers, and evidence-based treatment regimens for pa- tients with AAV. Epidemiology AAV is an infrequent disease with an incidence of about 20 per million population per year in North Amer- ica and Europe [6]. Incidence increases with age, with a peak incidence during the seventh decade of life and a marked predominance of male patients. AAV has a high- er prevalence in Caucasic and Asian populations than Af- rican-American populations [7]. There is notable geo- graphic variation, with GPA being more common in Northern Europe and Pacific area countries, whereas MPA is more common in Southern Europe and Asia [8]. Infection with Staphylococcus aureus may trigger epi- sodes of AAV since chronic nasal carriage of S. aureus has been associated with an increased risk for disease relapse [9]. Other environmental factors include silica exposure, hydrocarbon exposure, pesticides, and medications such as hydralazine and propylthiouracil [10–12]. Genetic Studies Two genome-wide association studies in North Amer- ican and European populations have identified disease susceptibility loci in AAV [4]. GPA is associated with sin- gle-nucleotide polymorphisms in HLA-DP, PRTN3 (en- coding PR3), and SERPINA1 (encoding α1-antitrypsin, a protease acting as the major inhibitor of PR3). PR3-AN- CA disease-associated variants in PRTN3 and SERPINA1 support the hypothesis that PR3-ANCA is not merely an epiphenomenon in AAV but plays a central role in the pathogenesisofthisdisease.Bycontrast,MPAwasassoci- atedwithHLA-DQpolymorphisms.Notably,thestrength of these genetic associations was greater with respect to ANCA specificity (PR3-ANCA or MPO-ANCA) than for the clinical phenotype (GPA or MPA) [13]. These data support for the concept that PR3-AAV and MPO-AAV are distinct autoimmune syndromes. Pathogenesis AAV pathogenesis has classically been focused on the activity and interactions of the neutrophil. They have a double role as the target of the ANCAs and effectors of the tissue damage after degranulation induced by these antibodies. The exposure of MPO and PR3 in the surface of the cellular membrane of the neutrophils is necessary for the binding of the ANCA and requires a pre-activa- tion process known as priming. Various microbial, en- dogenous, or environmental agents and pro-inflammato- ry cytokines such as interleukin (IL)-1, IL-18, and TNF-α can prime neutrophils. Once primed, neutrophils achieve their maximal phagocytic potential and response more intensively to a second stimulus such as ANCA binding. Proteolytic enzymes and free oxygen radicals are released from neutrophil granules into the damage site. Neutro- phils also release chemokines that recruit more neutro- phils and other inflammatory cells [14]. Moreover, MPO and PR3 can exert direct damage on endothelial cells [15]. In addition, neutrophils from AAV patients are more prone to NETosis, spontaneously or induced by ANCA, and tissular deposition of NETs also contributes to vas- cular endothelial damage [16] (Fig. 1). How ANCAs are produced is debated. MPO and PR3 are mostly circumscribed to the primary granules of the neutrophil’s cytoplasm and are rapidly eliminated by spe- cific inhibitors after neutrophil degranulation, thus stay- ing out of reach of the immune system [17]. Several hy- potheses have been drawn regarding how these proteins can become immunogenic, such as the molecular mim- icry with bacterial antigens hypothesis [18], the comple- mentary peptide hypothesis [19], or the exposure of MPO and PR3 as components of the NETs in NETosis process- es [16]. In recent years, this neutrophil-focused view of the AAV pathogenesis is being abandoned in favor of the consideration of the disease as a multistep process with
  • 3. ANCA Vasculitis: An Individualized Approach 123 Nephron 2022;146:121–137 DOI: 10.1159/000520727 the involvement of other immune cells. In this line, ex- tensive T-cell infiltrates have been identified in damaged tissue from AAV patients, as well as forming part of the characteristic granulomas [20]. Moreover, preclinical models of AAV are attenuated in the absence of different T-cell subsets, thus suggesting the importance of these cells for the induction of the disease [21]. The ameliora- tion of the disease after T-cell directed therapies is also a proof of concept of the T-cell involvement [22]. The aber- rant T-cell response in AAV consists on the polarization toward predominant Th1 [23] and Th17 [24] phenotypes during the acute phase of the disease, together with a nu- merical and functional impairment in T regulatory lym- phocytes [25]. Some authors described a higher propor- tion of activated CD8+ T cells in AAV patients together with a higher expression of IL-7 and T-cell receptor sig- naling pathways in these cells [26]. Altered T-cell co- stimulation has also been described in AAV, consisting of a decreased CD28 expression and CTLA4 overexpression with diminished inhibitory ability, thus resulting in resis- tance to anergy and contributing to a more pronounced pro-inflammatory state [27]. The participation of the alternate pathway of the complement has been demonstrated in AAV. Despite the paucity of the immune deposits in AAV lesions, el- evated serum C5a levels have been described in active AAV [28]. C5a acts as a powerful neutrophil priming agent via the C5a receptor present in this cell. At the same time, neutrophils release properdin that promotes further generation of C5a [29]. Based on this rationale, the utility of the C5a receptor inhibitor as induction to remission treatment in active AAV has recently been published [30]. Resting neutrophil Plasma leakage and fibrinoid necrosis Endothelial damage Chemotaxis of inflammatory cells Proteolytic enzymes release NETosis Direct ANCA damage ANCA binding Microbial, environmental or endogenous agents, proinflammatory cytokines, C5a complement fraction Primed neutrophil ROS release Molecular mimicry with bacterial antigens Immune response to complementary peptides Exposure of MPO and PR3 after netosis ANCA production Fig. 1. Pathophysiology. MPO and PR3 are inside cytoplasmic granules in resting neutrophils. Pre-activation (priming) of the neutrophils by environmental, infectious, or pro-inflammatory agents is required to expose MPO and PR3 in the cell surface. Once exposed, ANCAs can bind to MPO and PR3. ANCA binding leads to neutrophil degranulation that releases ROS and proteolytic en- zymes that cause endothelial damage. The rupture of the endothe- lium allows plasma leakage that causes fibrinoid necrosis. MPO, myeloperoxidase; PR3, proteinase 3; ANCA, antineutrophil cyto- plasmic antibody; ROS, reactive oxygen species.
  • 4. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 124 DOI: 10.1159/000520727 Clinical Presentation AAV is a heterogeneous group of autoimmune multi- system conditions that can present with a wide variety of signs and symptoms. Clinical manifestations depend on the size, number, and type of affected vessels and the or- gans involved, disease stage, and activity [5]. In addition to organ-specific manifestations, patients often present with constitutional symptoms such as fever, malaise, weight loss, anorexia, myalgias, and arthralgias which re- late to the systemic autoimmune pathophysiology [1]. The upper respiratory tract is usually compromised of de- structive inflammatory lesions causing nasal, sinus, tra- cheal, and/or ear abnormalities [31]. Upper respiratory tract and ear-nose-throat (ENT) involvement frequently is manifested by recurrent epistaxis, mucosal ulceration, nasal septum deformities and perforation, sinusitis, otitis media, or subglottic stenosis [32]. Pulmonary manifesta- tions of AAV include pulmonary nodules and diffuse al- veolar hemorrhage. Alveolar hemorrhage occurs as a re- sult of pulmonary capillaritis and presents with hemop- tysis and dyspnea. The most reported ocular involvement in AAV includes orbital disease and scleritis [33]. Cuta- neous [34], neurological [35], or enteric [36] involvement is also frequent (Table 1). Regarding to kidney involvement in AAV, it manifests as a rapidly progressive glomerulonephritis. Usually, it is accompanied by a nephritic syndrome with hematuria and proteinuria. Less frequently, renal involvement in AAV presents as subacute or chronic nephritis. Renal AAV can lead to end-stage renal disease. The different AAV disease phenotypes are composed of adifferentialpatternofassociationsoforgandamage.Prev- alence of renal involvement ranges between 75 and 90% and is higher in MPA than inGPA or EGPA [1]. Necrotiz- ing glomerulonephritis is present in >80% patients diag- nosed with MPA and up to 50% patients present with pul- monaryinvolvement.ThirtypercentofMPApatientspres- ent with neurological, skin, or gastrointestinal symptoms. In GPA patients, necrotizing vasculitis is accompanied by granulomatous inflammation. ENT and upper airway in- volvement are more frequent in GPA patients than in MPA patients. EGPA presents on a background of eosinophilia and asthma, and sinuses abnormalities are commonly as- sociated. Around one-third of patients have renal manifes- tations. Some patients with GPA or MPA present with vas- culitis limited to a single organ, such as the kidneys or lungs, which may represent the early stages of AAV. How- ever, in MPO-ANCA+ patients with MPA, isolated renal disease or isolated pulmonary fibrosis is not infrequent. Classification Criteria and Diagnosis The classification of the systemic vasculitis has been controversial for several decades. The American College of Rheumatology Criteria (ACR) was published in 1990 and included only GPA and EGPA but not MPA [37]. Lately, the Chapel Hill Consensus Conference (CHCC) definitions developed in 1994 and revised in 2012 added the knowledge of the etiopathogenesis of vasculitis. A new tree hierarchy was developed which recognized that some conditions cannot be simply classified by vessel size and suggested the use of surrogate markers of the disease, such as ANCAs. AAV was recognized as a specific type of small-vessel vasculitis along with immune complex-me- diated vasculitis (Fig. 2) [2]. In clinical practice, AAV serological diagnosis is based on the finding of ANCA positivity in sera from patients with consistent signs and symptoms. The presence of ANCA can be evaluated by means of the indirect immuno- fluorescence(IIF)testonhumanethanol-fixedneutrophils [38] by enzyme-linked immunoassay and more recently by chemiluminescent immunoassay. C-ANCA describes an IIF pattern consisting on a diffuse granular cytoplasmic staining predominantly associated to the presence of auto- antibodies targeting PR3. On the contrary, the IIF pattern P-ANCA consists on a perinuclear staining related to the presence of autoantibodies against MPO [39]. Results re- Table 1. Serological and clinical features of AAV GPA, % MPA, % EGPA, % PR3/C-ANCA 80–90 10–20 10–30 MPO/P-ANCA 10–15 70–80 40–60 Negative ANCA 5 10–20 10–50 Cutaneous 40 40 50 Kidney 80 90 40 Pulmonary 90 50 70 ENT 90 30 50 Neurological 50 30 70 Gastrointestinal 30–50 30–50 30–50 Musculoskeletal 60 60 50 Eyes 30 20 10 ANCA, antineutrophil cytoplasmic antibody; PR3, proteinase 3; MPO, myeloperoxidase; GPA, granulomatosis with polyangiitis; MPA, microscopic polyangiitis; EGPA, eosinophilic granulomatosis with polyangiitis; RLV, renal-limited vasculitis; ORL, otorhinolaryn- gological; AAV, antineutrophil cytoplasmic antibody-associated vasculitis; ENT, ear, nose, and throat.
  • 5. ANCA Vasculitis: An Individualized Approach 125 Nephron 2022;146:121–137 DOI: 10.1159/000520727 trieved from the IIF test, as a screening test, should be con- firmed by enzyme-linked immunoassay [40]. In the recent years, chemiluminescent immunoassay has been devel- oped in order to improve sensitivity and specificity in the detection and titration of ANCAs [39]. Regarding the clinical syndrome, MPA-AAV patients often show a P-ANCA pattern associated to the presence of anti-MPO antibodies [41], but they can also exhibit anti-PR3 antibodies. The IIF pattern most commonly as- sociated with GPA, but not exclusive, is C-ANCA, related to anti-PR3 antibodies [42]. In EGPA, <50% of patients have detectable ANCAs, more frequently anti-MPO an- tibodies [43]. More recently, results from epidemiological outcomes and genetic studies in AAV suggest that patients should be classified by ANCA specificity rather by the clinical syn- drome [4]. The new Diagnostic and Classification Criteria of Vasculitis Study aims to provide new validated classifi- cation and diagnostic criteria for systemic vasculitis [44]. Disease Activity and Damage Scores Validated tools to assess disease activity are Birming- ham Vasculitis Activity Score (BVAS) and the Five-Fac- tor Score. The BVAS includes 10 categories of new or worsening symptoms presented <4 weeks until the re- cord. A BVAS score of 0 represents remission, ≥1 repre- sents active disease, and refractory disease is active dis- ease despite treatment [45]. The 2009 Five-Factor Score for EGPA comprises serum creatinine, age (>65 years), cardiomyopathy, gastrointestinal involvement, and the absence of ENT manifestations [46]. On the other hand, the Vasculitis Damage Index is used to assess chronic damage from the disease and treatment and includes musculoskeletal, skin, and mucous membranes and ocu- lar, ENT, pulmonary, cardiovascular, the peripheral vas- culature, gastrointestinal, renal, and neuropsychiatric systems, with an eleventh category for other systems [47]. Biomarkers There is a rising interest in the use of biomarkers in screening disease, diagnosing, staging, monitoring thera- peutic interventions, predicting outcomes or adverse events, or identifying cell types in AAV. Although con- troversial, the most accepted classical biomarker of active AAV is the ANCA titer. In a subgroup of 104 AAV pa- tients with renal involvement, Kemna et al. [48] found a correlation between ANCA titer rise and disease relapse. Lionaki et al. [49] described that ANCA-PR3 specificity Large vessel Takayasu arteritis Giant cell arteritis Medium vessel Polyarteritis nodosa Kawasaki disease Cryoglobulinaemic vasculitis lgA vasculitis Hypocomplementemic urticarial vasculitis Anti-GBM disease ANCA-associated vasculitis Small vessel Fig. 2. Classification of AAV. AAV, antineutrophil cytoplasmic antibody-associated vasculitis; Ig, immunoglobulin.
  • 6. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 126 DOI: 10.1159/000520727 predicts relapse in AAV patients with kidney involve- ment, conferring almost twice the relapse risk compared to ANCA-MPO vasculitis. Based on the disease pathophysiology, neutrophil ac- tivation biomarkers are of special interest in AAV. After ANCA binding and full activation of these cells, neutro- phil microparticles and NETs are released and can be measured in the serum of AAV patients for the assess- ment of disease activity [50, 51]. Calprotectin is contained in the cytoplasmic granules of neutrophils and released after degranulation in active AAV. Calprotectin binds en- dothelial cells and amplifies endothelial damage and in- flammation, by increasing IL8, ICAM1, and leukocyte re- cruitment [52]. Pepper et al. [52] demonstrated calpro- tectin expression by immunohistochemistry in kidney biopsies from AAV patients localized in crescents and ar- eas of endocapillary proliferation. They also found that serum calprotectin is elevated in patients in the acute phase of AAV compared to patients in remission [53]. Our group described higher serum calprotectin levels in remission in those patients with worse renal outcomes during the follow-up and found higher urinary calprotec- tin levels in the acute patients than in the remission pa- tients [54]. Also, in the field of urinary biomarkers, suCD163 has shown promising results. suCD163 is pro- duced by macrophages composing the inflammatory in- filtrates in AAV, released to the urinary space, and mea- sured in the urine. Higher suCD163 levels have been no- ticedinacuteAAVwithrenalinvolvementthanremission [55]. In a prospective study, suCD163 consistently in- creased compared to previous levels during the follow-up of patients in remission who experienced relapse [56]. suCD163 has been evaluated in combination with sCD25 [57] or MCP1 [58], showing an improved performance as a disease activity biomarker. Due to their emerging role in disease pathogenesis, some T-cell subpopulations or byproducts have been de- scribed as AAV biomarkers. Our group found higher fre- quencies of MPO/PR3-specific Th17 cells in acute AAV than remission and suggested higher specific Th17 count in remission as a surrogate marker of subclinical activity [59]. Morgan et al. [60] found that patients with a higher Treg count at presentation entered earlier in remission than those with a lower Treg count and a negative corre- lation between Tregs and the disease relapse rate. In the same line, Yoshimura et al. [61] correlated a lower FOXP3 staining – a key transcription factor for the suppressive ability of Tregs – in AAV renal biopsies with the require- ment of maintenance. McKinney et al. [26] found that the higher expression of a particular subgroup of genes re- lated to T-cell survival, and expansion of memory CD8 T cells was strongly associated with a shorter time to relapse after induction to remission treatment and identified those individuals with a greater propensity for relapse, al- lowing the customization of the therapy. Inthesettingoftherisinginterestinthealternatepath- way of the complement as a therapeutic target in AAV, complement fractions have also been studied as biomark- ers. Villacorta et al. observed that serum C3 at baseline was predictive of renal and global survival in AAV pa- tients, with the worse outcomes associated with C3 con- sumption [62] and that C3d-positive staining in diagnos- tic kidney biopsy was associated with the severity of renal impairment and with a lower response rate to treatment [63]. Treatment Remission Induction Therapy Advances in immunosuppressive treatment have dras- tically reduced mortality of AAV in the past decades. Re- mission rates obtained in different clinical trials range be- tween 53 and 90% [64] (Table  2). However, mortality rates despite immunosuppressive treatment are not neg- ligible, especially in those patients presenting with severe organ involvement, such as renal failure or alveolar hem- orrhage [65]. Infection constitutes the major cause of death within the first year after the onset, followed by ac- tive vasculitis damage. Therefore, AAV therapy should try to control disease activity but minimize side-effect de- velopment, mainly infections. The goal of induction therapy is to achieve remission within the first 3 months, by employing more intensive immunosuppression. Before initiation of therapy, careful assessment of disease activity, organ damage, and quality of life is crucial and may guide the use of potentially tox- ic therapies. Remission has been defined as improvement or stability of renal function, resolution of hematuria, and the absence of extrarenal features of disease activity. Treatment should be initiated as soon as AAV with organ and life-threatening manifestations are suspected, even in the absence of histological confirmation of vasculitis since a delay in treatment can lead to worse outcomes. CYC-Based Remission Induction Treatment AAV with organ and life-threatening manifestations has been classically treated with combination therapy of glucocorticoids and CYC for 3–6 months. CYC can be given orally or intravenously. Both routes of administra-
  • 7. ANCA Vasculitis: An Individualized Approach 127 Nephron 2022;146:121–137 DOI: 10.1159/000520727 Table 2. Induction immunosuppressive protocol according to different trials Study Drug comparison Induction immunosuppressive protocol Maintenance therapy Results immunosuppressive drug doses and duration corticosteroid dose corticosteroid tapering CYCLOPS RCT (2009) ivCYC versus oral CYC ivCYC 15 mg/kg every 2–3 weeks for 3–6 months Oral prednisolone 1 mg/kg Tapering to 12.5 mg/ day at month 3 and to 5 mg at month 18 Oral AZA 2 mg/kg/day since month 3–6 to month 18 No differences in remission rates. Less cumulative dose in the ivCYC group Oral CYC 2 mg/kg/day for 3–6 months RAVE RCT (2010) RTX versus oral CYC RTX 375 mg/m 2 /week for 4 weeks 1–3 pulses 1 g iv methylprednisolone, followed by oral prednisolone at 1 mg/kg/day Tapering to zero at month 5 Placebo maintenance RTX noninferior to oral CYC for induction remission RTX superior for relapsing vasculitis or PR3-positive patients Oral CYC 2 mg/kg/day for 3–6 months Oral AZA 2 mg/kg/day since month 3–6 to month 18 RITUXVAS RCT (2010) RTX versus ivCYC RTX + 2 doses ivCYC±PEX/ SDS RTX 375 mg/m 2 /week for 4 weeks + CYC 15 mg/kg with the first and third RTX 1 pulse 1 g iv methylprednisolone, followed by oral prednisolone at 1 mg/kg/day Tapering to 5 mg/day at month 6 Withdrawn at month 12 No maintenance RTX noninferior to ivCYC for induction remission ivCYC 15 mg/kg every 2 weeks for 3–6 months Oral AZA 2 mg/kg/day since month 3–6 to month 24 Pepper et al. [86] Cohort study (2019) LDS versus SDS from historic cohort of EUVAS trials RTX + ivCYC RTX 1 g at day 0 and day 7 CYC 500 mg, 6 doses every 2 weeks 2 pulses 250–500 mg methylprednisolone at days 0 and 7, followed by 0.5 mg/kg/day (max 30 mg) days 2–6 Withdrawn at day 7 (maximum duration 3 weeks) Oral AZA 2 mg/kg/day No differences in remission rates RTX 1 g at day 0 and day 14 CYC 500 mg, 6 doses every 2 weeks 1 pulse 250–1,000 mg methylprednisolone, followed by 60 mg/day 1 week, followed by 45 mg/day 1 week Withdrawn at week 2 (5 patients received >30 days of prednisolone) MYCYC RCT (2019) Oral MMF versus ivCYC Oral MMF 2 g/day until remission Oral prednisolone 1 mg/kg Tapering to 5 mg/day at month 6 Prednisolone 5 mg/day + oral AZA 2 mg/kg/day since remission to month 18 No differences in remission rates At 18 months: more relapses with MMF, especially in PR3- ANCA patients IvCYC 15 mg/kg every 2–3 weeks until remission CYCLOWVAS Cohort study (2019) LDS versus SDS from historic cohort of EUVAS trials RTX + ivCYC RTX 1 g at day 0 and day 14 CYC 10 mg/kg to maximum of 500 mg, 6 doses every 2 weeks Oral prednisolone 1 mg/kg (maximum dose 60 mg) Tapering to 10 mg/day at week 13 Oral AZA 2 mg/kg/day since month 3 to month 18–24 High rate remission Reduced risk of death, ESRD and relapses compared with the historic cohort PEXIVAS RCT (2020) LDS versus SDS RTX/ivCYC±PEX RTX/ivCYC according to local practitioner PEX according to the PEXIVAS trial randomization 1–3 pulses 1 g iv methylprednisolone, followed by oral prednisolone at 1 mg/kg/day Tapering to 5 mg/day at week 23–52 No maintenance specified No differences in remission rates between groups 1–3 pulses 1 g methylprednisolone, followed by oral prednisolone at 1 mg/kg/day Tapering to 5 mg/day at week 15–16 ADVOCATE RCT (2021) Avacopan versus prednisone RTX + ivCYC RTX (375 mg/m 2 weekly for 4 weeks) or CYC orally (2 mg/ kg daily for 14 weeks) or IV (15 mg/kg every 2–3 weeks for 13 weeks Prednisone: 60 mg versus avacopan: 30 mg orally twice daily for 52 weeks without prednisone Taper over 21 weeks Oral AZA 2 mg/kg/day since month 3–6 to month 18 after CYC The RTX arm did not receive maintenance At weeks 26 and 52: Noninferior remission rate in the avacopan group compared to prednisone CYC, cyclophosphamide; ivCYC, intravenous cyclophosphamide; AZA, azathioprine; RTX, rituximab; MMF, mycophenolate mofetil; PEX, plasma exchange; SDS, standard dose of steroids; LDS, low dose of steroids; EUVAS, European Renal Association and the European Vasculitis Society.
  • 8. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 128 DOI: 10.1159/000520727 tion were evaluated in the CYCLOPS trial, in which pa- tients with a new diagnosis of AAV were randomly as- signed to receive either oral CYC or intravenous CYC [66]. There was no difference in remission rates between the 2 groups (88.1% vs. 87.7%), but the cumulative dose was significantly higher in the oral CYC group (15.2 vs. 8.2 g) and was associated with a higher incidence of leu- kopenia. Long-term follow-up of the trial showed that al- though the oral CYC group was at lower risk for relapse, this did not translate into differences in renal or overall survival [67]. For this reason, several clinical guidelines recommend intravenous CYC as induction therapy [67, 68]. CYC is associated with several serious adverse effects such as infection, bone marrow suppression, hemorrhag- ic cystitis, and infertility [68]. Several studies have linked CYC therapy with an increased incidence of malignances, mainly skin cancer, myeloid malignancies, and bladder cancer [69, 70]. This increased risk for the development of tumors is especially evident when the cumulative dose is >20 gr [71]. Therefore, induction treatment with CYC should be avoided in patients with cancer history, those with high risk or active infection, and young patients with gestational desire. Rituximab-Based Remission Induction Treatment Given the substantial toxicity associated with cumula- tive CYC use and the relapsing nature of AAV, the use of rituximab (RTX), a chimeric anti-CD20 monoclonal an- tibody, has been widely employed as induction therapy [71]. Two randomized controlled trials, RAVE and RITUXVAS, evaluated the efficacy of RTX for remission induction in GPA and MPA [72, 73]. In the RAVE trial, mainly composed by PR3-ANCA patients with both new and relapsing diseases, but without severe renal failure, the RTX arm received methylprednisolone pulses, and the prednisone dosage was tapered to zero within 6 months. On the other hand, the RITUXVAS trial enrolled only patients with newly diagnosed vasculitis with more severe kidney disease, including patients requiring dialy- sis (20%). In contrast to the RAVE trial, the RTX arm re- ceived 2 doses of intravenous CYC, and the use of plas- mapheresis was allowed. Both studies demonstrated that RTX therapy was noninferior to CYC for remission in- duction, with comparable rates of mortality and adverse events. A subanalysis of the RAVE trial performed among patients with relapsing disease concluded that RTX was superior to CYC in PR3-ANCA patients with relapsing disease since a higher rate of remission in the RTX arm was observed [74]. Based on this, current guidelines rec- ommend RTX as first-line treatment for PR3-ANCA pa- tients, relapsing disease, refractory disease, and those with contraindications to CYC [75, 76] (Fig. 3). Specific measures such as prevention of hepatitis B vi- rus reactivation and hypogammaglobulinemia surveil- lance must be taken when receiving RTX therapy [77]. Up to 25% of patients with AAV present hypogammaglobu- linemia at the onset of AAV disease and after RTX ther- apy low levels of immunoglobulin (Ig) G are observed in up to 50–60% of patients [78]. In most cases, hypogam- maglobulinemia is mild and transient, and IgG levels re- turntonormalwithoutspecificmeasureswithin6months after RTX infusion. However, severe hypogammaglobu- linemia occurs in 4.2% of patients and contributes to re- current infections, requiring the use of intravenous Ig. Therefore, Ig levels should be checked at baseline and at least before each RTX infusion [79]. Individuals with chronic hepatitis B and previously in- fected but serologically cleared HBV infection are both susceptible to HBV reactivation after RTX therapy [80]. Identification of patients at risk and institution of pro- phylactic antiviral therapy prior to initiation of RTX is essential [81]. Finally, late-onset neutropenia is a rare side effect of RTX observed among AAV patients that mani- fests as abrupt and severe neutropenia within 2–6 months following the last dose of RTX and may require filgrastim administration in some cases [82]. Mycophenolate-Based Remission Induction Treatment The use of mycophenolate mofetil (MMF) for induc- tion remission therapy was evaluated in a randomized controlled trial (MYCYC) which compared intravenous CYC versus oral MMF (2–3 g/d) for treatment of newly diagnosed AAV patients without severe kidney failure [83]. The remission rates observed at 6 months were com- parable between both groups (67% vs. 61%). However, relapses were more common in the MMF group, mostly in PR3-ANCA-positive patients. Therefore, the study suggests that MMF and glucocorticoids can be used as a first-line induction therapy, mainly in patients with MPO-ANCA who have mild to moderate renal involve- ment without life-threatening extrarenal vasculitis. Novel Strategies for Remission Induction Therapy: Steroid Minimization Regimens Classically, oral glucocorticoids have constituted a cornerstone of AAV induction therapy due to its rapid effect which allows disease control in the meanwhile oth- er immunosuppressives to have an effect. However, glu- cocorticoids have an extensive side-effect profile, includ-
  • 9. ANCA Vasculitis: An Individualized Approach 129 Nephron 2022;146:121–137 DOI: 10.1159/000520727 ing hypertension, hyperglycemia, weight gain, bone dis- ease, psychiatric disorders, gastrointestinal bleeding, and long-term risks for cardiovascular disease [84]. Although intravenous pulse methylprednisolone at the onset of therapy for severe disease is conventionally administered, its benefits have not been adequately stud- ied. In recent years, several studies have explored the em- ploy of lower dose regimens of steroids for induction therapy and similar efficacy with a reduced risk of severe infections was observed. The CYCLOWVAS study pro- posed a combination of RTX with quinquennial low-dose pulses of CYC associated with a rapid taper of glucocor- ticoids within the first month [85]. A high remission rate was observed in this study and when compared with an European Renal Association and the European Vasculitis Society (EUVAS) historic cohort, a reduction of the risk of progression to end-stage renal disease and death was shown. Another observational study employing this im- munosuppression protocol represented a step forward toward glucocorticoid withdrawal [86]. In this study, prednisone was discontinued within the first 2 weeks of therapy. Patients with severe renal failure were not ex- cluded, and however, the remission rates observed were very high (>90%). The rapid withdrawal of prednisone was associated with a significant lower incidence of car- diovascular events and diabetes. However, further trials are needed to confirm the safety and superiority of this combination regimen. Finally, the recent PEXIVAS trial demonstrated that a reduction of the prednisone dose to 20 mg daily by 7 weeks and 5 mg daily by 20 weeks was as Mild to moderate kidney failure New onset MPO PR3/MPO Relapsed disease Severe kidney failure New onset/relapsed disease PR3/MPO RTX + SDS a) RTX + ivCYC + LDS (*) e) RTX + ivCYC + SDS g) RTX + ivCYC + LDS (*) h) PR3 RTX + SDS a)/LDS d) No remission Remission No remission No remission No remission Consider stop treatment and/or dialysis Swich therapy Remission Remission Maintenance therapy RTX 500 mg every 4–6 months i) (+) AZA j) ivCYC ivCYC RTX + SDS a)/LDS d) ivCYC + SDS b)/LDS d) MMF + SDS c) RTX + iv CYC + LDS (*) e) Avacopan + RTX/ivCYC f) Fig. 3. Treatment algorithm for AAV. Mild to moderate kidney failure: GFR >15 mL/min/1.73 m2 ; severe kidney failure: GFR <15 mL/min/1.73 m2 . a) RAVE RCT protocol; b) CYCLOPS RCT pro- tocol; c) MYCYC RCT protocol; d) PEXIVAS trial; e) CYCLOW- VAS case-control trial protocol (RTX + quinquennial low-dose CYC); f) AVACOPAN RCT protocol; g) RITUXVAS RCT proto- col (RTX + quinquennial low-dose CYC); h) case control study protocol from Pepper et al. [86]; i) MAINRITSAN 1 RCT protocol; j) CYCAZREM RCT protocol. (*) Patients with intolerance or contraindication to corticosteroids (+) or according to ANCA sta- tus or CD19 B-cell repopulation (MAINRITSAN 2 protocol). Maintenance for 24 months in patients with low risk of relapse and up to 48 months in PR3-ANCA patients and those with high risk for relapsing. PR3, proteinase 3; MPO, myeloperoxidase; RTX, rituximab; SDS, standard dose of steroids; LDS, low dose of ste- roids; ivCYC, intravenous cyclophosphamide; MMF, mycopheno- late mofetil; AZA, azathioprine; RCT, randomized clinical trial; AAV, antineutrophil cytoplasmic antibody-associated vasculitis; CYC, cyclophosphamide.
  • 10. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 130 DOI: 10.1159/000520727 effective as a higher dose regimen and implied a substan- tial reduction in the infection rates [87]. Complement Blockage: Betting on the Future Given the knowledge of the crucial role of complement activation in the pathogenesis of AAV, therapies target- ing the alternative pathway and particularly the anaphyl- atoxin C5a have emerged [28]. The ADVOCATE trial was a phase 3 study which analyzed standard immuno- suppression with CYC or RTX combined with a selective C5a receptor inhibitor avacopan or oral prednisone on a tapering schedule [30]. At week 26, the proportion of pa- tients in disease remission (BVAS = 0) in the avacopan group was comparable to the prednisone group (72.3% vs. 70.1%) [29]. Sustained remission at week 52 was ob- served in 65.7% of the patients receiving avacopan and in 54.9% receiving prednisone. Therefore, avacopan nonin- feriority was met at weeks 26 and 52 (p < 0.001), and su- periority was met at week 52 only (p = 0.007). Avacopan patients reported better health-related quality of life in several measures (physical health, health perception, emotional health, and vitality). However, the study had some limitations. First, the large number of patients in the avacopan arm (86%) who received nonstudy-supplied glucocorticoids within the first 6 months may have had an impact on the study drug effect. Second, the lack of maintenance therapy in the RTX subgroup may also have had some positive impact outcome for the avacopan arm since the difference for long-term disease remission was only observed among patients treated with RTX. Finally, similar infection rates were observed in both groups of treatment, but a greater proportion of avacopan-treated patients had adverse events associated with hepatic abnormalities. Despite these concerns, the avacopan groups were noninferior to the standard prednisone group in the study, highlighting a potential approach in which glucocorticoid doses can be minimized. Another phase II study is currently analyzing the employ of vilobelimab (IFX-1), a first-in-class anti- C5a antibody, added to standard of care and will certain- ly provide further information about complement inhibi- tion in AAV induction therapy [88]. Plasma Exchange: Unlearning What Has Been Learned? The benefit for considering plasma exchange (PEX) in treatment of patients AAV is that the removal of ANCAs and other inflammatory mediators could promote earlier reversal of the immunological response and minimize tis- sue damage. However, PEX is an invasive therapeutic method which could increase the risk of infection, hem- orrhage, and catheter-related complications and when frozen plasma is employed, transfusion-related adverse reactions [89]. The MEPEX trial showed that adjuvant therapywithPEXaddedtoCYCinAAVpatientspresent- ing with severe kidney failure was associated with a 24% risk reduction in progression to end-stage kidney disease [90]. However, the longer term follow-up data showed no difference in mortality or end-stage kidney disease [91]. The PEXIVAS enrolled a total of 704 MPO and PR3-AAV patients with renal involvement and with or without pul- monary compromise [87]. Treatment with PEX associ- ated to standard immunosuppression (RTX or CYC) was compared with no PEX, and a standard dose oral gluco- corticoid regimen was compared with a reduced dose reg- imen. Notably, serious adverse events, including serious infections, were comparable among patients treated with and without PEX but were less frequent among patients receiving a low dose of steroids. After a median follow-up of 3 years, the study showed no difference in primary end points (death from any cause or kidney failure) between groups receiving or not PEX, and subgroup analysis failed to show a benefit among those patients requiring dialysis at the onset (20%). However, similarly to the MEPEX study, the analysis at the 1-year follow-up showed a nonsignificant advan- tage of PEX (HR, 0.77; 95% CI, 0.56–1.06), whereas sta- tistical assessment at 3 and 6 months was not provided despite short-term clear separation between survival curves. Therefore, many nephrologists consider that a short-term benefit of PEX leading to a free dialysis period within the first year, as well as the absence of serious tech- nique-related complications in the study, still justifies PEX employ among patients presenting with severe renal failure. In the PEXIVAS trial, 61 (9%) patients with severe pul- monary hemorrhage, defined as oxygen saturation ≤85% or requirement of mechanical ventilation, were included. Although PEX did not show a statistically significant ef- fect on the primary outcome in these patients, a trend toward the beneficial effect of PEX could be observed (HR,0.67;95%CI,0.28–1.6).However,theimpactofPEX on mortality alone was not provided, and the study had not powerful enough to evaluate the true effect of PEX in these patients. Maintenance Therapy: Which One and for How Long? AAV constitutes a chronic disease with a relapsing course. Up to 30–60% of patients will present a relapse in the course of the disease, mainly within the first 2 years
  • 11. ANCA Vasculitis: An Individualized Approach 131 Nephron 2022;146:121–137 DOI: 10.1159/000520727 Table 3. Maintenance immunosuppressive protocol according to different trials Study Immunosuppressive drug induction Maintenance comparison Maintenance drug Doses and duration Steroid maintenance Results CYCAZAREM RCT (2003) Oral CYC + SDS Oral CYC versus AZA Oral CYC 1.5 mg/kg/day until month 12, followed by AZA 1.5 mg/kg/day Prednisolone 10 mg/day until month 12, then tapering to 7.5 mg/day No differences in relapse rates AZA 2 mg/kg/day until month 12, then tapering to 1.5 mg/kg/day IMPROVE RCT (2010) CYC + SDS MMF versus AZA MMF 2 g/kg/day, tapering to 1.5 g/kg/day after 12 months, 1 g/kg/day after 18 months, withdrawn after 42 months Prednisolone 15 mg/day at the start of maintenance, tapering to 5 mg/day after 12 months, withdrawn after 24 months Higher relapse rates in the MMF group AZA 2 mg/kg/day, tapering to 1.5 mg/kg/day after 12 months, 1 mg/kg/day after 18 months, withdrawn after 42 months MAINRITSAN RCT (2014) ivCYC + SDS RTX versus AZA RTX 500 mg at days 0 and 14, then at months 6, 12, and 18 Prednisone tapering to 5 mg/day until 18 months, then withdrawn according to physician criteria Greater sustained remission in the RTX group AZA 2 mg/kg/day for 12 months, 1.5 mg/kg/ day for 6 months, 1 mg/kg/day for 4 months REMAIN RCT (2017) CYC + SDS AZA + LDS for 24 months versus AZA + LDS for 48 months AZA + LDS for 24 months Withdrawn AZA at month 24 Withdrawn steroids at month 24 Greater sustained remission in the prolonged AZA group AZA + LDS for 48 months AZA 1 mg/kg/day until month 48 Tapering prednisolone from 5 to 7.5 mg/day to zero at month 48 MAINRITSAN 2 RCT (2018) CYC + SDS Fixed RTX dosing versus tailored RTX dosing Fixed 500 mg IV at days 0 and 14 and then at 6, 12, and 18 months No significant difference in the relapse rate. Less RTX infusions in the tailored group Individualized 500 mg IV at randomization and then reinfusion only if reappearance of CD19 or ANCA or increased titer of ANCA Prednisone tapering to 5 mg/day until 18 months, then withdrawn according to physician criteria MAINRITSAN 3 RCT (2020) CYF/MTX/RTX + SDS* RXT for 18 months (5 doses) versus RTX for 36 months (9 doses) RTX 18 months 500 mg at day 0 and 14, then at months 6, 12, and 18 Prednisone tapering to 5 mg/day until 18 months, then withdrawn according to physician criteria Greater sustained remission in the prolonged RTX group RTX 46 months 500 mg at days 0 and 14, then at months 6, 12, and 18, then at months 28, 34, 40, and 46 CYC, cyclophosphamide; AZA, azathioprine; RTX, rituximab; MMF, mycophenolate mofetil; MTX, methotrexate; SDS, standard dose of steroids; LDS, low dose of steroids. *Patients in the MAINRITSAN 2 trial.
  • 12. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 132 DOI: 10.1159/000520727 after the withdrawal of immunosuppression. The appear- ance of relapses implies a substantial increase in the mor- bidity in these patients. Risk factors for relapse include PR3-ANCA serotype, GPA phenotype, ENT involve- ment, persistent ANCA-positive titers, and persistent mi- crohematuria [86, 92]. The use of maintenance immunosuppression for re- lapse prevention following successful remission induc- tion is standard (Table 3). The first study to compare 2 maintenance regimens was the CYCAZAREM trial, in which patients with AAV who achieved remission with oral CYC were randomly assigned at 6 months to con- tinue CYC therapy or switch to azathioprine (AZA) [93]. At 18 months, relapse rates were similar between both arms (15.5 vs. 13.7%) but with less side effects in the AZA group. The IMPROVE trial compared MMF to AZA and showed higher rates of relapse in patients treated with MMF. Therefore, MMF may be considered for remission maintenance in patients who have intolerance to AZA. Subsequently, the employ of RTX as maintenance therapy showed dramatically lower rates of relapse. In the MAINRITSAN 1 trial, patients who achieved remission after CYC induction regimen were randomized to receive either AZA or biannual semestral RTX infusions (500 mg) as maintenance therapy [94]. In the RTX arm, only 5% of treated patients experienced a major relapse com- pared to 29% in the AZA group. This beneficial effect of RTX in the prevention of relapses was independent of the ANCA serotype, and no difference in side effects or infec- tion rates was observed between groups. The ongoing RITAZAREM trial analyzes another RTX maintenance regimen (1 g every 4 months for 2 years) with conven- tional maintenance therapy with AZA in patients with relapsing AAV who received RTX for induction. The first report of the trial showed a high level of efficacy (90%) with RTX in conjunction with glucocorticoids for the re- induction of remission in patients with AAV who have relapsed [95]. An approach to individualized mainte- nance therapy employing RTX dosing tailored to B-cell reappearance and/or increase in ANCA titers was evalu- ated in the MAINRITSAN 2 trial [96]. In this study, the median number of infusions was reduced (3 vs. 5) in the tailored-dosegroupcomparedtopatientsreceivingfixed- dose RTX every 6 months. Although not significant, a trend toward a higher rate of relapse in patients with RTX-tailored dosing (17.3%) compared with patients re- ceiving fixed-dose RTX (9.9%) was observed. Interesting- ly, 50% of the relapses in this study occurred in patients without B-lymphocyte reconstitution and 30% with neg- ative ANCA, highlighting the limitation of these bio- markers to predict relapse. Notably, up to 30% of patients experienced a relapse 38 months after the last RTX infu- sion, putting into debate the optimal duration of mainte- nance immunosuppression. In this sense, the REMAIN trial was a randomized con- trolled trial that tested whether continuing AZA and prednisone treatment for 48 months was more effective in relapse prevention than withdrawal at 24 months [97]. Results from this study showed a significant decrease in both major and minor relapses and better renal survival in the continuation group. More recently, the MAINRIT- SAN 3 trial included PR3 and MPO-AAV patients who were in complete remission after receiving an 18-month RTX maintenance regimen in the MAINRITSAN 2 study [98], and participants were randomly assigned to receive prolonged maintenance therapy with either intravenous RTX (500 mg every 6 months) or a placebo for a further 18 months. Relapse-free survival rates at month 28 were significantly higher among patients given prolonged RTX (96%) than patients given the placebo (74%), and long- term RTX maintenance therapy did not increase the number of adverse events. Notably, relapses occurred more frequently in PR3-ANCA patients than in MPO pa- tients in the placebo arm (40% vs. 12%). On the basis of these results, prolonged maintenance treatment (4–5 years) with RTX may be employed at high-risk patients for relapses, such as those with PR3-ANCA, those who have already had a relapse, or with persistent hematuria or high ANCA titers despite treatment. Refractory Disease and Other Therapies Refractory disease in AAV can be defined as progres- sive disease within the first 3 months or lack control of vasculitis activity by 6 months despite standard of care therapy. An increase in the glucocorticoid dose is used in severe disease, but prolonged use of high-dose oral gluco- corticoids should be avoided due to the associated risks. Switching from CYC to RTX and vice versa or a combina- tion of both immunosuppressants could be considered in refractory cases. The development of anti-RTX antibodies has been de- scribed to neutralize RTX B-cell cytotoxicity and may im- pact the clinical outcome of autoimmune diseases [99]. Ofatumumab, a fully humanized antibody directed against a distinct extracellular epitope of CD20 has slow- er dissociation kinetics than that of RTX and has been shown to be a more potent activator of complement-de- pendent cytotoxicity in vitro. It was successfully em- ployed in small series of AAV cases [100], but its role in refractory AAV disease requires further investigation.
  • 13. ANCA Vasculitis: An Individualized Approach 133 Nephron 2022;146:121–137 DOI: 10.1159/000520727 The efficacy and safety of mepolizumab, a human monoclonal antibody against IL-5, for the treatment of relapsing or refractory EGPA, were investigated in a ran- domized, placebo-controlled study which included pa- tients on a stable corticosteroid therapy [101]. Patients treated with mepolizumab achieved a significantly higher rate of remission than patients who received the placebo. Also beneficial effects of IL-6 receptor blockade therapy with tocilizumab in association with steroids have been observed in small series of MPA patients [102], but larger and prospective studies are desirable to confirm these re- sults. Given the considerable evidence for the role of TNF-α in pathogenesis of AAV, several anti-TNF-α agents have been explored for AAV therapy. The WGET study was a randomized, placebo-controlled study which examined the use of etanercept as add-on therapy to conventional treatment, for maintenance of remission, in a large cohort of patients with GPA [103]. This study found no addi- tional benefit of etanercept, and excess of solid cancers was observed, but this risk could not be attributed solely to etanercept treatment since previous history of malig- nancy and CYC exposure previous to etanercept could have also played a role. Another anti-TNF agent, inflix- imab, was evaluated as adjuvant therapy with CYC and corticosteroids in a prospective clinical trial [104]. Inflix- imab was effective at inducing remission in 88% of pa- tients and permitted reduction in steroid doses. However, severe infections were seen in 21% of patients, and despite continued infliximab, 20% of initial responders experi- enceddiseaseflares.Adalimumabwassimilarlyemployed as adjuvant therapy showing high rates of early remission (80% at 3 months) and the ability to significantly reduce the steroid dose during induction treatment [105]. Other therapeutic approaches include the combina- tion of the BLyS/BAFF blockade with RTX, which is now being evaluated in the “Rituximab and Belimumab Com- bination Therapy in PR3 Vasculitis” trial [106]. Alemtu- zumab, a humanized anti-CD52 (CAMPATH-1H) monoclonal antibody, was evaluated in combination with prednisone for patients with AAV refractory to conven- tional treatment [107]. In this study, 85% of patients achieved remission, but 71% of these had relapsed by 9.2 months and adverse events were common. Leflunomide was reported efficacious for remission induction among 62 out of 93 patients (67%) with differ- ent vasculitic disorders (64% with GCA, 89% with TAK, 80% with PAN, 69% with GPA, 75% with MPA, and 33% with EGPA) [108]. In this study, 20% of patients discon- tinued leflunomide before achieving remission because of persistent disease activity and adverse events (gastroin- testinal symptoms being the most common) led to drug discontinuation in 18 (19%) patients. Antithymocyte globulin (ATG) was also employed as adjuvant therapy in a small cohort of patients with refrac- tory GPA [109]. The majority of patients achieved remis- sion after therapy, and treatment with ATG allowed a fur- ther reduction in immunosuppression. However, authors recommended avoiding ATG therapy if simultaneous in- fections, fluid overload, or alveolar hemorrhage were present. Also in GPA patients with refractory disease, a 6-month course of the immunosuppressant 15-deoxys- pergualin was successfully employed in 7 patients [110]. However, most patients relapsed after 15-deoxyspergua- lin discontinuation. There is little experience with agents targeting plasma cells in AAV. Experimental studies showed that bortezo- mib depleted MPO-specific plasma cells and prevented anti-MPO IgG-mediated necrotizing crescentic glomeru- lonephritis in mice [111]. Interestingly, a favorable effect of proteasome inhibition was reported in 1 patient with AAV [112]. Finally, the use of the Ig was analyzed in a randomized, placebo-controlled trial that investigated the efficacy of a single course of intravenous Ig (IVIg) (total dose 2 g/kg) in previously treated AAV patients, showing persistent disease activity [113]. A single course of IVIg reduced dis- ease activity in these patients, but the effect was not main- tained beyond 3 months. Therefore, IVIg may be an alter- native treatment option for AAV with persistent disease activity after standard therapy. In this sense, the ongoing Endurance 1 trial is a randomized and prospective study which aims to prove the superiority of combination treat- ment RTX with CYC compared to standard of care ther- apy in AAV patients showing persistent disease activity [114]. Conclusions AAVs have a complex pathogenesis, influenced by ge- netics and environmental factors. There is evidence for a loss of tolerance to neutrophil proteins, which leads to ANCA-mediated neutrophil activation, recruitment, and injury, with effector T cells and activation of alternative pathway of complement also involved. NETs containing MPO and PR3 contribute to the pathogenesis through different mechanisms, including antigen presentation that promotes autoimmunity as well as direct vascular damage.
  • 14. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 134 DOI: 10.1159/000520727 Data from epidemiological outcomes and genetic studies in AAV suggest that patients should be classified by ANCA specificity (MPO-ANCA vs. PR3-ANCA) rath- er than by the clinical syndrome. Novel biomarkers for screening disease, monitoring therapeutic interventions and identifying relapses include neutrophil activation markers such as calprotectin, macrophage activation-de- rived products (uCD163 and MCP), T-cell subpopula- tions, and alternative pathway of complement fractions. The treatment of AAV is rapidly evolving, and mortal- ity and remission rates have improved over the years. However, high infection rates are still observed, and cur- rent strategies are focused on reducing treatment toxicity. RTX-based regimens are the cornerstone for induction therapy among PR3 and relapsing patients as well as for maintenance therapy. Novel therapeutic strategies in- clude blockage of complement and combination of im- munosuppressants, which allow minimization of cumu- lative steroids exposure. Individualization of treatment according to ANCA specificity, disease phenotype, and risk of relapse is a key for offering a personalized care to AAV patients. Acknowledgments We thank all the members of the Spanish Group for the Study of Glomerular Diseases (GLOSEN) for their valuable contribution to research in glomerular diseases. Conflict of Interest Statement The authors have no conflicts of interest to declare. Funding Sources There were no funding sources for this work. Author Contributions All the authors contributed equally to this study, revised the paper, and approved the final version of the manuscript. References   1 Rowaiye OO, Kusztal M, Klinger M. The kid- neys and ANCA-associated vasculitis: from pathogenesis to diagnosis. Clin Kidney J. 2015 Jun;8(3):343–50.   2 Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, et al. 2012 revised Interna- tional Chapel Hill Consensus Conference No- menclature of Vasculitides. Arthritis Rheum. 2013 Jan;65(1):1–11.   3 Jennette JC, Nachman PH. ANCA glomeru- lonephritis and vasculitis. Clin J Am Soc Nephrol. 2017 Oct;12(10):1680–91.  4 Lyons PA, Rayner TF, Trivedi S, Holle JU, Watts RA, Jayne DRW, et al. Genetically dis- tinct subsets within ANCA-associated vascu- litis. N Engl J Med. 2012 Jul;367(3):214–23.   5 Yates M, Watts R. ANCA-associated vasculi- tis. Clin Med. 2017 Feb;17(1):60.   6 Berti A, Dejaco C. Update on the epidemiol- ogy, risk factors, and outcomes of systemic vasculitides. Best Pract Res Clin Rheumatol. 2018;32(2):271–94.   7 Watts RA, Robson J. Introduction, epidemi- ology and classification of vasculitis. Best Pract Res Clin Rheumatol. 2018;32(1):3–20.   8 Fujimoto S, Watts RA, Kobayashi S, Suzuki K, Jayne DRW, Scott DGI, et al. Comparison of the epidemiology of anti-neutrophil cytoplas- mic antibody-associated vasculitis between Japan and the U.K. Rheumatology. 2011 Oct; 50(10):1916–20.   9 Popa ER, Stegeman CA, Abdulahad WH, van der Meer B, Arends J, Manson WM, et al. Staphylococcal toxic-shock-syndrome-tox- in-1 as a risk factor for disease relapse in We- gener’s granulomatosis. Rheumatology. 2007 Jun;46(6):1029–33. 10 Gómez-Puerta JA, Gedmintas L, Costenbader KH. The association between silica exposure and development of ANCA-associated vascu- litis: systematic review and meta-analysis. Autoimmun Rev. 2013 Oct;12(12):1129–35. 11 Clarkson AR, Robson JS. The effect of drugs on immunological renal disease. Prog Bio- chem Pharmacol. 1972;7:427–51. 12 Draibe J, Rodó X, Fulladosa X, Martínez-Va- lenzuelaL,Diaz-EncarnaciónM,SantosL,etal. Seasonal variations in the onset of positive and negative renal ANCA-associated vasculitis in Spain. Clin Kidney J. 2018 Aug;11(4):468–73. 13 Rahmattulla C, Mooyaart AL, van Hooven D, Schoones JW, Bruijn JA, Dekkers OM, et al. Genetic variants in ANCA-associated vascu- litis: a meta-analysis. Ann Rheum Dis. 2016 Sep;75(9):1687–92. 14 Mayadas TN, Cullere X, Lowell CA. The mul- tifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218. 15 Wilde B, Thewissen M, Damoiseaux J, van Paassen P, Witzke O, Tervaert JW. T cells in ANCA-associated vasculitis: what can we learn from lesional versus circulating T cells? Arthritis Res Ther. 2010;12(1):204. 16 Kessenbrock K, Krumbholz M, Schöner- marck U, Back W, Gross WL, Werb Z, et al. Netting neutrophils in autoimmune small- vessel vasculitis. Nat Med. 2009 Jun;15(6): 623–5. 17 Aratani Y. Myeloperoxidase: its role for host defense, inflammation, and neutrophil func- tion. Arch Biochem Biophys. 2018 Feb;640: 47–52. 18 Kain R, Exner M, Brandes R, Ziebermayr R, Cunningham D, Alderson CA, et al. Molecu- lar mimicry in pauci-immune focal necrotiz- ing glomerulonephritis. Nat Med. 2008 Oct; 14(10):1088–96. 19 Pendergraft WF, Preston GA, Shah RR, Trop- sha A, Carter CW, Jennette JC, et al. Autoim- munity is triggered by cPR-3(105-201), a pro- tein complementary to human autoantigen proteinase-3. Nat Med. 2004 Jan;10(1):72–9. 20 Balding CE, Howie AJ, Drake-Lee AB, Savage CO. Th2 dominance in nasal mucosa in pa- tients with Wegener’s granulomatosis. Clin Exp Immunol. 2001 Aug;125(2):332–9. 21 Ruth AJ, Kitching AR, Kwan RYQ, Odobasic D, Ooi JDK, Timoshanko JR, et al. Anti-neu- trophil cytoplasmic antibodies and effector CD4+ cells play nonredundant roles in anti- myeloperoxidase crescentic glomerulone- phritis. J Am Soc Nephrol. 2006 Jun;17(7): 1940–9. 22 Walsh M, Chaudhry A, Jayne D. Long-term follow-up of relapsing/refractory anti-neu- trophil cytoplasm antibody associated vascu- litis treated with the lymphocyte depleting an- tibody alemtuzumab (CAMPATH-1H). Ann Rheum Dis. 2007 Nov;67(9):1322–7.
  • 15. ANCA Vasculitis: An Individualized Approach 135 Nephron 2022;146:121–137 DOI: 10.1159/000520727 23 Abdulahad WH, van der Geld YM, Stegeman CA, Kallenberg CG. Persistent expansion of CD4+ effector memory T cells in Wegener’s granulomatosis. Kidney Int. 2006 Sep;70(5): 938–47. 24 Abdulahad WH, Stegeman CA, Limburg PC, Kallenberg CG. Skewed distribution of Th17 lymphocytes in patients with Wegener’s gran- ulomatosis in remission. Arthritis Rheum. 2008 Jul;58(7):2196–205. 25 Reijnders TDY, Stegeman CA, Huitema MG, Rutgers A, Heeringa P, Abdulahad WH. Un- raveling the identity of FoxP3+ regulatory T cells in granulomatosis with polyangiitis pa- tients. Sci Rep. 2019;9(1):8273–9. 26 McKinney EF, Lyons PA, Carr EJ, Hollis JL, Jayne DRW, Willcocks LC, et al. A CD8+ T cell transcription signature predicts progno- sis in autoimmune disease. Nat Med. 2010 May;16(5):586–91. 27 Moosig F, Csernok E, Wang G, Gross WL. Costimulatory molecules in Wegener’s gran- ulomatosis (WG): lack of expression of CD28 and preferential up-regulation of its ligands B7-1 (CD80) and B7-2 (CD86) on T cells. Clin Exp Immunol. 1998 Oct;114(1):113–8. 28 Moiseev S, Lee JM, Zykova A, Bulanov N, Novikov P, Gitel E, et al. The alternative com- plement pathway in ANCA-associated vascu- litis: further evidence and a meta-analysis. Clin Exp Immunol. 2020 Dec;202(3):394– 402. 29 Geetha D, Jefferson JA. ANCA-associated vasculitis: core curriculum 2020. Am J Kidney Dis. 2020 Jan;75(1):124–37. 30 Jayne DRW, Merkel PA, Schall TJ, Bekker P. Avacopan for the treatment of ANCA-associ- ated vasculitis. N Engl J Med. 2021 Feb; 384(7):599–609. 31 Said MS. Upper respiratory tract symptoms, renal involvement and vasculitis: a case report and review of wegener granulomatosis. J Clin Med Res. 2010 Aug;2(4):189–93. 32 Parra-García GD, Callejas-Rubio JL, Ríos- Fernández R, Sainz-Quevedo M, Ortego- Centeno N. Otolaryngologic manifestations of systemic vasculitis. Acta Otorrinolaringol. 2012 Jul;63(4):303–10. 33 Watkins AS, Kempen JH, Choi D, Liesegang TL, Pujari SS, Newcomb C, et al. Ocular dis- ease in patients with ANCA-positive vasculi- tis. J Ocul Biol Dis Inform. 2009 Dec;3(1): 12–9. 34 Demirkesen C. Approach to cutaneous vascu- litides with special emphasis on small vessel vasculitis: histopathology and direct immu- nofluorescence. Curr Op Rheumatol. 2017 Jan;29(1):39–44. 35 Wludarczyk A, Szczeklik W. Neurological manifestations in ANCA-associated vasculi- tis: assessment and treatment. Expert Rev Neurother. 2016 Aug;16(8):861–3. 36 Storesund B, Gran JT, Koldingsnes W. Severe intestinal involvement in Wegener’s granulo- matosis: report of two cases and review of the literature. Br J Rheumatol. 1998 Apr;37(4): 387–90. 37 Hunder GG, Arend WP, Bloch DA, Calabrese LH, Fauci AS, Fries JF, et al. The American College of Rheumatology 1990 criteria for the classification of vasculitis: introduction. Ar- thritis Rheum. 1990;33(8):1065–7. 38 Rasmussen N, Wiik A. Indirect immunofluo- rescence examination for IgG-ANCA in sera submitted for the 1st international workshop on ANCA, 1988. APMIS Suppl. 1989;6:16– 20. 39 Schulte-Pelkum J, Radice A, Norman GL, Lόpez Hoyos M, Lakos G, Buchner C, et al. Novel clinical and diagnostic aspects of anti- neutrophil cytoplasmic antibodies. J Immu- nol Res. 2014;2014:185416. 40 Savige J, Gillis D, Benson E, Davies D, Esnault V, Falk RJ, et al. International consensus statement on testing and reporting of anti- neutrophil cytoplasmic antibodies (ANCA). Am J Clin Pathol. 1999 Apr;111(4):507–13. 41 Chung SA, Seo P. Microscopic polyangiitis. Rheum Dis Clin North Am. 2010 Aug;36(3): 545–58. 42 Lutalo PMK, D’Cruz DP. Diagnosis and clas- sification of granulomatosis with polyangiitis (aka Wegener’s granulomatosis). J Autoim- mun. 2014 Feb;48:94–8. 43 Gioffredi A, Maritati F, Oliva E, Buzio C. Eo- sinophilic granulomatosis with polyangiitis: an overview. Front Immunol. 2014;5:549. 44 Yates M, Watts R, Bajema I, Cid M, Crestani B, Hauser T, et al. Validation of the EULAR/ ERA-EDTA recommendations for the man- agement of ANCA-associated vasculitis by disease content experts. RMD open. 2017 Jun; 3(1):e000449. 45 Mukhtyar C, Lee R, Brown D, Carruthers D, Dasgupta B, Dubey S, et al. Modification and validation of the birmingham vasculitis activ- ity score (version 3). Ann Rheum Dis. 2009 Dec;68(12):1827–32. 46 Guillevin L, Pagnoux C, Seror R, Mahr A, Mouthon L, Toumelin PL. The five-factor score revisited: assessment of prognoses of systemic necrotizing vasculitides based on the french vasculitis study group (FVSG) cohort. Medicine. 2011 Jan;90(1):19–27. 47 Exley AR, Bacon PA, Luqmani RA, Kitas GD, Gordon C, Savage COS, et al. Development and initial validation of the vasculitis damage index for the standardized clinical assessment of damage in the systemic vasculitides. Ar- thritis Rheumat. 1997;40(2):371–80. 48 Kemna MJ, Damoiseaux J, Austen J, Winkens B, Peters J, van Paassen P, et al. ANCA as a predictor of relapse: useful in patients with re- nal involvement but not in patients with non- renal disease. J Am Soc Nephrol. 2015 Mar; 26(3):537–42. 49 Lionaki S, Blyth ER, Hogan SL, Hu Y, Senior BA, Jennette CE, et al. Classification of anti- neutrophil cytoplasmic autoantibody vasculi- tides: the role of antineutrophil cytoplasmic autoantibody specificity for myeloperoxidase or proteinase 3 in disease recognition and prognosis. Arthritis Rheumat. 2012 Oct; 64(10):3452–62. 50 Hong Y, Eleftheriou D, Hussain AAK, Price- Kuehne FE, Savage CO, Jayne D, et al. Anti- neutrophil cytoplasmic antibodies stimulate release of neutrophil microparticles. J Am Soc Nephrol. 2012;23:49–62. 51 Wang H, Sha LL, Ma TT, Zhang LX, Chen M, Zhao MH. Circulating level of neutrophil ex- tracellular traps is not a useful biomarker for assessing disease activity in antineutrophil cy- toplasmic antibody-associated vasculitis. PLoS One. 2016 Feb 3;11(2):e0148197. 52 Pepper RJ, Wang HH, Rajakaruna GK, Pa- pakrivopoulou E, Vogl T, Pusey CD, et al. S100A8/A9 (calprotectin) is critical for devel- opment of glomerulonephritis and promotes inflammatory leukocyte-renal cell interac- tions. Am J Pathol. 2015 May;185(5):1264– 74. 53 Pepper RJ, Hamour S, Chavele KM, Todd SK, Rasmussen N, Flint S, et al. Leukocyte and se- rum S100A8/S100A9 expression reflects dis- ease activity in ANCA-associated vasculitis and glomerulonephritis. Kidney Int. 2013 Jun;83(6):1150–8. 54 Martinez Valenzuela L, Draibe J, Ramos MQ, Fulladosa Oliverasid X, Melilli E, Cruzado Garrit JM, et al. Calprotectin as a smoldering activity detection tool and renal prognosis biomarker in ANCA associated vasculitis. PLoS One. 2018;13(10). 55 O’Reilly VP, Wong L, Kennedy C, Elliot LA, O’Meachair S, Coughlan AM, et al. Urinary soluble CD163 in active renal vasculitis. J Am Soc Nephrol. 2016 Sep;27(9):2906–16. 56 Villacorta J, Lucientes L, Goicoechea E, Ace- vedo M, Cavero T, Sanchez-Camara L, et al. Urinary soluble CD163 as a biomarker of disease activity and relapse in antineutro- phil cytoplasm antibody-associated glomer- ulonephritis. Clin Kidney J. 2021 Feb;14(1): 212–9. 57 Dekkema GJ, Abdulahad WH, Bijma T, Mo- ran SM, Ryan L, Little MA, et al. Urinary and serum soluble CD25 complements urinary soluble CD163 to detect active renal anti-neu- trophil cytoplasmic autoantibody-associated vasculitis: a cohort study. Nephrol Dial Transplant. 2019 Feb;34(2):234–24. 58 Moran SM, Monach PA, Zgaga L, Cuthbert- son D, Carette S, Khalidi NA, et al. Urinary soluble CD163 and monocyte chemoattrac- tant protein-1 in the identification of subtle renal flare in anti-neutrophil cytoplasmic an- tibody–associated vasculitis. Nephrol Dial Transplant. 2020 Feb;35(2):283–91. 59 Valenzuela LM, Draibe J, Quero M, Fulladosa X, Cruzado JM, Bestard O, et al. Exploring frequencies of circulating specific th17 cells against myeloperoxidase and proteinase 3 in anca associated vasculitis. Int J Mol Sci. 2019 Dec;20(23):5820. 60 Morgan MD, Day CJ, Piper KP, Khan N, Harper L, Moss PA, et al. Patients with We- gener’s granulomatosis demonstrate a rela- tive deficiency and functional impairment of T-regulatory cells. Immunology. 2010 May; 130(1):64–73.
  • 16. Villacorta/Martinez-Valenzuela/ Martin-Capon/Bordignon-Draibe Nephron 2022;146:121–137 136 DOI: 10.1159/000520727 61 Yoshimura J, Fukami K, Koike K, Nagano M, Matsumoto T, Iwatani R, et al. Interstitial Foxp3-positive T cells may predict renal sur- vival in patients with myeroperoxidase anti- neutrophil cytoplasmic antibody-associated glomerulonephritis. Clin Exp Pharmacol Physiol. 2010 Sep;37(9):879–83. 62 Villacorta J, Diaz-Crespo F, Acevedo M, Cavero T, Guerrero C, Praga M, et al. Circu- lating C3 levels predict renal and global out- come in patients with renal vasculitis. Clin Rheumatol. 2016 Nov;35(11):2733–40. 63 Villacorta J, Diaz-Crespo F, Acevedo M, Guerrero C, Campos-Martin Y, García-Díaz E, et al. Glomerular C3d as a novel prognostic marker for renal vasculitis. Human Pathol. 2016 Oct;56:31–9. 64 Robson J, Doll H, Suppiah R, Flossmann O, Harper L, Höglund P, et al. Damage in the an- ca-associated vasculitides: long-term data from the European vasculitis study group (EUVAS) therapeutic trials. Ann Rheum Dis. 2015 Jan;74(1):177–84. 65 Flossmann O, Berden A, de Groot K, Hagen C, Harper L, Heijl C, et al. Long-term patient survival in ANCA-associated vasculitis. Ann Rheum Dis. 2011 Mar;70(3):488–94. 66 de Groot K, Harper L, Jayne DRW, Flores Su- arez LF, Gregorini G, Gross WL, et al. Pulse versus daily oral cyclophosphamide for in- duction of remission in antineutrophil cyto- plasmic antibody-associated vasculitis: a ran- domized trial. Ann Intern Med. 2009 May; 150(10):670–80. 67 Harper L, Morgan MD, Walsh M, Hoglund P, Westman K, Flossmann O, et al. Pulse versus daily oral cyclophosphamide for induction of remission in ANCA-associated vasculitis: long-term follow-up. Ann Rheum Dis. 2012 Jun;71(6):955–60. 68 Wester Trejo MAC, Bajema IM, van Daalen EE. Antineutrophil cytoplasmic antibody-as- sociated vasculitis and malignancy. Curr Opin Rheumatol. 2018 Jan;30(1):44–9. 69 Lafarge A, Joseph A, Pagnoux C, Puechal X, Cohen P, Samson M, et al. Risk of malignancy in patients treated for systemic necrotising vasculitis. Ann Rheum Dis. 2020 Mar;79(3): 431–3. 70 Knight A, Askling J, Granath F, Sparen P, Ek- bom A. Urinary bladder cancer in Wegener’s granulomatosis: risks and relation to cyclo- phosphamide. Ann Rheum Dis. 2004 Oct; 63(10):1307–11. 71 Emadi A, Jones RJ, Brodsky RA. Cyclophos- phamide and cancer: golden anniversary. Nat Rev Clin Oncol. 2009 Nov;6(11):638–47. 72 Jones RB, Tervaert JWC, Hauser T, Luqmani R, Morgan MD, Peh CA, et al. Rituximab ver- sus cyclophosphamide in ANCA-associated renal vasculitis. N Engl J Med. 2010;363(3): 211–20. 73 Stone JH, Merkel PA, Spiera R, Seo P, Lang- ford CA, Hoffman GS, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. 2010 Jul;363(3): 221–32. 74 Unizony S, Villarreal M, Miloslavsky EM, Lu N, Merkel PA, Spiera R, et al. Clinical out- comes of treatment of anti-neutrophil cyto- plasmic antibody (ANCA)-associated vascu- litis based on ANCA type. Ann Rheum Dis. 2016 Jun;75(6):1166–9. 75 Yates M, Watts RA, Bajema IM, Cid MC, Crestani B, Hauser T, et al. EULAR/ERA-ED- TA recommendations for the management of ANCA-associatedvasculitis.AnnRheumDis. 2016 Sep;75(9):1583–94. 76 Rovin BH, Caster DJ, Cattran DC, Gibson KL, Hogan JJ, Moeller MJ, et al. Management and treatment of glomerular diseases (part 2): conclusions from a Kidney Disease: Improv- ing Global Outcomes (KDIGO) Controver- sies Conference. Kidney Int. 2019 Feb;95(2): 281–95. 77 McClure M, Gopaluni S, Jayne D, Jones R. B cell therapy in ANCA-associated vasculitis: current and emerging treatment options. Nat Rev Rheumatol. 2018 Oct;14(10):580–91. 78 Roberts DM, Jones RB, Smith RM, Alberici F, Kumaratne DS, Burns S, et al. Rituximab-as- sociated hypogammaglobulinemia: inci- dence, predictors and outcomes in patients with multi-system autoimmune disease. J Au- toimmun. 2015 Feb;57:60–5. 79 Tieu J, Smith R, Basu N, Brogan P, D’Cruz D, Dhaun N, et al. Rituximab for maintenance of remission in ANCA-associated vasculitis: ex- pert consensus guidelines. Rheumatology. 2020 Apr;59(4):e24–32. 80 Tsutsumi Y, Yamamoto Y, Ito S, Ohigashi H, Shiratori S, Naruse H, et al. Hepatitis B virus reactivation with a rituximab-containing reg- imen. World J Hepatol. 2015 Sep;7(21):2344– 51. 81 Loomba R, Liang TJ. Hepatitis B reactivation associated with immune suppressive and bio- logical modifier therapies: current concepts, management strategies, and future directions. Gastroenterology. 2017 May;152(6):1297–309. 82 Zonozi R, Wallace ZS, Laliberte K, Huizenga NR, Rosenthal JM, Rhee EP, et al. Incidence, clinical features, and outcomes of late-onset neutropenia from rituximab for autoimmune disease. Arthritis Rheumatol. 2021 Feb;73(2): 347–54. 83 Jones RB, Hiemstra TF, Ballarin J, Blockmans DE, Brogan P, Bruchfeld A, et al. Mycopheno- late mofetil versus cyclophosphamide for re- mission induction in ANCA-associated vas- culitis: a randomised, non-inferiority trial. Ann Rheum Dis. 2019 Mar;78(3):399–405. 84 Grennan D, Wang S. Steroid side effects. JAMA. 2019 Jul;322(3):282. 85 McAdoo SP, Medjeral-Thomas N, Gopaluni S, Tanna A, Mansfield N, Galliford J, et al. Long-term follow-up of a combined ritux- imab and cyclophosphamide regimen in renal anti-neutrophil cytoplasm antibody-associat- ed vasculitis. Nephrol Dial Transplant. 2019 Jan;34(1):63–73. 86 Pepper RJ, McAdoo SP, Moran SM, Kelly D, Scott J, Hamour S, et al. A novel glucocorti- coid-free maintenance regimen for anti-neu- trophil cytoplasm antibody-associated vascu- litis. Rheumatology. 2019 Feb;58(2):373. 87 Walsh M, Merkel PA, Peh CA, Szpirt WM, Puéchal X, Fujimoto S, et al. Plasma exchange and glucocorticoids in severe ANCA-associ- ated vasculitis. N Engl J Med. 2020 Feb; 382(7):622–31. 88 U.S National Library of Medicine. Safety and Efficacy Study of IFX-1 in add-on to standard of care in granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA). 2021. Available from: https://clinicaltrials. gov/ct2/show/NCT03712345. 89 Lu J, Zhang L, Xia C, Tao Y. Complications of therapeutic plasma exchange: a retrospective study of 1201 procedures in 435 children. Medicine. 2019 Dec;98(50):e18308. 90 Jayne DRW, Gaskin G, Rasmussen N, Abramowicz D, Ferrario F, Guillevin L, et al. Randomized trial of plasma exchange or high-dosage methylprednisolone as adjunc- tive therapy for severe renal vasculitis. J Am Soc Nephrol. 2007 Jul;18(7):2180–8. 91 Walsh M, Casian A, Flossmann O, Westman K, Höglund P, Pusey C, et al. Long-term fol- low-up of patients with severe ANCA-associ- ated vasculitis comparing plasma exchange to intravenous methylprednisolone treatment is unclear. Kidney Int. 2013 Aug;84(2):397–402. 92 Salama AD. Relapse in anti-neutrophil cyto- plasm antibody (ANCA)–associated vasculi- tis. Kidney Int Rep. 2019 Oct 24;5(1):7–12. 93 Jayne D, Rasmussen N, Andrassy K, Bacon P, Tervaert JWC, Dadoniené J, et al. A random- ized trial of maintenance therapy for vasculi- tis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med. 2003 Jul; 349(1):36–44. 94 Guillevin L, Pagnoux C, Karras A, Khouatra C, Aumaître O, Cohen P, et al. Rituximab ver- sus azathioprine for maintenance in ANCA- associated vasculitis. N Engl J Med. 2014 Nov; 371(19):1771–80. 95 Smith RM, Jones RB, Specks U, Bond S, No- dale M, Aljayyousi R, et al. Rituximab as ther- apy to induce remission after relapse in AN- CA-associated vasculitis. Ann Rheum Dis. 2020 Sep;79(9):1243–9. 96 Charles P, Terrier B, Perrodeau É, Cohen P, Faguer S, Huart A, et al. Comparison of indi- vidually tailored versus fixed-schedule ritux- imab regimen to maintain ANCA-associated vasculitis remission: results of a multicentre, randomised controlled, phase III trial (MAINRITSAN2). Ann Rheum Dis. 2018 Aug;77(8):1143–9. 97 Karras A, Pagnoux C, Haubitz M, Groot K, Puechal X, Tervaert JWC, et al. Randomised controlled trial of prolonged treatment in the remission phase of ANCA-associated vasculi- tis. Ann Rheum Dis. 2017 Oct;76(10):1662–8. 98 Charles P, Perrodeau É, Samson M, Bonnotte B, Néel A, Agard C, et al. Long-term ritux- imab use to maintain remission of antineu- trophil cytoplasmic antibody-associated vas- culitis: a randomized trial. Ann Intern Med. 2020 Aug;173(3):179–87.
  • 17. ANCA Vasculitis: An Individualized Approach 137 Nephron 2022;146:121–137 DOI: 10.1159/000520727  99 Boyer-Suavet S, Andreani M, Lateb M, Savenkoff B, Brglez V, Benzaken S, et al. Neutralizing anti-rituximab antibodies and relapseinmembranousnephropathytreated with rituximab. Front Immunol. 2019;10: 3069. 100 McAdoo SP, Bedi R, Tarzi R, Griffith M, Pusey CD, Cairns TD. Ofatumumab for B cell depletion therapy in ANCA-associated vasculitis: a single-centre case series. Rheu- matology. 2016 Aug;55(8):1437–42. 101 Wechsler ME, Akuthota P, Jayne D, Khoury P,KlionA,LangfordCA,etal.Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis. N Engl J Med. 2017; 376(20):1921–32. 102 Sakai R, Kondo T, Kurasawa T, Nishi E, Okuyama A, Chino K, et al. Current clinical evidence of tocilizumab for the treatment of ANCA-associated vasculitis: a prospective case series for microscopic polyangiitis in a combination with corticosteroids and litera- ture review. Clin Rheumatol. 2017 Oct; 36(10):2383–92. 103 The Wegener’s Granulomatosis Etanercept Trial (WGET) Research Group. Etanercept in addition to standard therapy in patients with Wegener’s granulomatosis. N Engl J Med. 2005;352:351–61. 104 Booth A, Harper L, Hammad T, Bacon P, Griffith M, Levy J, et al. Prospective study of TNF-ablockadewithinfliximabinanti-neu- trophil cytoplasmic antibody-associated systemic vasculitis. J Am Soc Nephrol. 2004; 15:717–72. 105 Laurino S, Chaudhry A, Booth A, Conte G, Jayne D. Prospective study of TNFalpha blockade with adalimumab in ANCA-asso- ciated systemic vasculitis with renal involve- ment. Nephrol Dial Transplant. 2010 Oct; 25(10):3307–14. 106 U.S National Library of Medicine. Ritux- imab and belimumab combination therapy in PR3 vasculitis (COMBIVAS). 2020. Available from: https://clinicaltrials.gov/ ct2/show/NCT03967925. 107 Walsh M, Chaudhry A, Jayne D. Long- term follow-up of relapsing/refractory an- tineutrophil cytoplasm antibody associat- ed vasculitis treated with the lymphocyte depleting antibody alemtuzumab (CAM- PATH-1H). Ann Rheum Dis. 2008;67: 1322–7. 108 Mustapha N, Barra L, Carette S, Cuthbert- son D, Khalidi NA, Koening CL, et al. Cana- dian vasculitis research network (CanVasc) and vasculitis clinical research consortium (VCRC). Efficacy of leflunomide in the treatment of vasculitis. Clin Exp Rheumatol. 2021;129(2):114–8. 109 Schmitt WH, Hagen EC, Neumann I, Nowack R, Flores-Suárez LF, Van der WoudeFJ,etal.TreatmentofrefractoryWe- gener’s granulomatosis with antithymocyte globulin (ATG): an open study in 15 pa- tients. Kidney Int. 2004;65:1440–8. 110 Flossmann O, Jayne DR. Long-term treat- ment of relapsing Wegener’s granulomato- sis with 15-deoxyspergualin. Rheumatology. 2010 Mar;49(3):556–62. 111 Bontscho J, Schreiber A, Manz RA, Schnei- der W, Luft FC, Kettritz R. Myeloperoxi- dase-specific plasma cell depletion by bortezomib protects from anti-neutrophil cytoplasmic autoantibodies-induced glo- merulonephritis. J Am Soc Nephrol. 2011 Feb;22(2):336–48. 112 Novikov P, Moiseev S, Bulanov N, Shchegol- eva E. Bortezomib in refractory ANCA-as- sociated vasculitis: a new option? Ann Rheum Dis. 2016 Jan;75(1):e9. 113 Jayne DR, Chapel H, Adu D, Misbah S, O’Donoghue D, Scott D, et al. Intravenous immunoglobulin for ANCA-associated sys- temic vasculitis with persistent disease activ- ity. QJM. 2000 Jul;93(7):433–9. 114 U.S National Library of Medicine. Exploring durable remission with rituximab in anti- neutrophil cytoplasmic antibody(ANCA)- associated vasculitis (ENDURRANCE-1). 2021. Available from: https://clinicaltrials. gov/ct2/show/NCT03942887?term= ENDURRANCE&draw=2&rank=1.