Skip to main content
European Journal of Rheumatology logoLink to European Journal of Rheumatology
. 2022 Jan 24;9(3):153–166. doi: 10.5152/eujrheum.2022.20248

Updates in ANCA-associated vasculitis

Carolyn Ross 1,, Jean-Paul Makhzoum 2,, Christian Pagnoux 1,
PMCID: PMC12462881  PMID: 35156630

Abstract

Antineutrophil cytoplasm antibody (ANCA)-associated vasculitides (AAV) are small-vessel vasculitides that include granulomatosis with polyangiitis (formerly Wegener’s granulomatosis), microscopic polyangiitis, and eosinophilic granulomatosis with polyangiitis (Churg - Strauss syndrome). Renal-limited AAV can be considered a fourth entity. Despite their rarity and still unknown cause(s), research into AAV has been very active over the past decades and has allowed for the development of new therapeutic regimens. The pathogenesis is a complex process of immune dysregulations with genetic and environmental influences. Recent genome-wide association studies have identified multiple genetic predisposing variants, especially at the major histocompatibility complex region. The pathogenic role of antimyeloperoxidase ANCA (MPO-ANCA) is well supported by several animal models, but that of antiproteinase 3 ANCA (PR3-ANCA) is not as strongly demonstrated. B cells likely play a major role in the pathogenesis because they produce ANCAs, as do neutrophil abnormalities, imbalances in T-cell subtypes, and/or cytokine - chemokine networks. The role of the alternative complement pathway was established more recently, and studies of the antagonist of human C5a receptor (avacopan) in AAV have just been completed, with promising results. The current standard management of severe AAV still consists of remission induction therapy with glucocorticoids combined with rituximab or, less often now, cyclophosphamide. Several studies showed that reduced-dose regimens of glucocorticoids are noninferior to the previously used heavier regimens, for therefore less cumulative exposure to glucocorticoids. Avacopan use may even lead to new steroid-free therapeutic approaches, at least for some selected patients. Several trials and studies have now shown the superiority of rituximab over azathioprine or methotrexate as maintenance therapy. However, the optimal dosing regimen and duration for maintenance remain to be better defined, at the individual patient level. Many changes have occurred in the standard of care for AAV over the past decades, and more are expected soon, including with use of avacopan, but also, likely, a few other agents under investigation or development.

Keywords: Anti-neutrophil cytoplasm antibody-associated vasculitides, granulomatosis with polyangiitis, microscopic polyangiitis, eosinophilic granulomatosis with polyangiitis, cyclophosphamide


Main Points

  • Despite their rarity and still unknown cause(s), research into AAV has been very active over the past decades and has allowed for the development of new therapeutic regimens.

  • Management of severe GPA and MPA still consists of remission induction therapy with glucocorticoids combined with rituximab or, less often now, cyclophosphamide.

  • Several studies have now shown superiority of rituximab over azathioprine or methotrexate as maintenance therapy in GPA and MPA.

  • More changes are expected soon, including with use of avacopan, the antagonist of human C5a receptor (avacopan), which may allow new steroidfree therapeutic approaches in GPA and MPA.

  • Treatment of EGPA also consists of a remission induction therapy followed by a maintenance phase. Mepolizumab, an anti-IL-5 humanized monoclonal antibody, showed clinical benefit, especially for the frequent steroid-dependent asthma or ear-nose-throat manifestations.

Introduction

Antineutrophil cytoplasm antibody (ANCA)-associated vasculitides (AAV) are small-vessel vasculitides that include granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), and eosinophilic granulomatosis with polyangiitis (EGPA) (Table 1).14 Renal-limited ANCA-associated vasculitis can be considered a fourth entity and is characterized by pauci-immune crescentic glomerulonephritis without systemic involvement and, in most cases, is an ANCA-positive disease with myeloperoxidase (MPO)-ANCA in 70%80% of patients.1 Despite the rarity and still unknown cause(s) of AAV, research into these diseases has been very active and steadily increasing over the past three decades. The results of several clinical and more basic fundamental studies demonstrated that each of these diseases has some different pathogenic mechanisms and genetic backgrounds.5 From a therapeutic point of view, treatment strategies have been gradually better defined, and several targeted biologic agents, initially developed for other diseases, have been studied for AAVs and/or are still under investigation.69 As demonstrated in two randomized controlled trials, the monoclonal anti-CD20 antibody rituximab has become an alternative to the conventional cytotoxic cyclophosphamide to induce remission in adults with severe GPA or MPA, combined with glucocorticoids.1012 Later, rituximab was also found more effective than azathioprine in maintaining disease remission.13 Other agents more recently studied to optimize treatment options in AAV include avacopan, an oral antagonist of human C5a receptor (C5aR), which could become the alternative to glucocorticoids in AAV, and belimumab, with more mitigated, nuanced results for maintaining remission in GPA. This article provides a brief overview of AAV characteristics and summarizes the results of some of the main recently published studies of AAV that may impact practice.

Table 1.

Classification criteria and definitions of the antineutrophil cytoplasm antibody (ANCA)-associated vasculitides according to the American College of Rheumatology (ACR, 1990; microscopic polyangiitis was not yet individualized as a specific entity at that time), and the 2012 Chapel Hill nomenclature.13

1990 ACR classification criteria for Wegener’s granulomatosis
 For purposes of classification, a patient shall be said to have Wegener’s granulomatosis if at least two of these four criteria are present. The presence of any two or more criteria yields a sensitivity of 88.2% and a specificity of 92.0 %.
  1. Nasal or oral inflammation: Development of painful or painless oral ulcers or purulent or bloody nasal discharge.

  2. Abnormal chest radiograph: Chest radiograph showing the presence of nodules, fixed infiltrates, or cavities.

  3. Urinary sediment: Microhematuria (>5 red blood cells per high power field) or red cell casts in urine sediment.

  4. Granulomatous inflammation on biopsy: Histologic changes showing granulomatous inflammation within the wall of an artery or in the perivascular or extravascular area (artery or arteriole).

1990 ACR classification criteria for Churg–Strauss syndrome
 For purposes of classification, a patient shall be said to have Churg–Strauss syndrome if at least four of these six criteria are present. The presence of any four or more criteria yields a sensitivity of 85% and a specificity of 99.7 %.
  1. Asthma: History of wheezing or diffuse high-pitched expiratory rhonchi.

  2. Eosinophilia greater than 10% on differential white blood cell count.

  3. Mononeuropathy (including multiplex) or polyneuropathy: Development of mononeuropathy, multiple mononeuropathies, or polyneuropathy (glove/ stocking distribution) attributable to systemic vasculitis.

  4. Nonfixed pulmonary infiltrates: Migratory or transitory pulmonary infiltrates (not including fixed infiltrates) attributable to vasculitis.

  5. Paranasal sinus abnormality: History of acute or chronic paranasal sinus pain or tenderness or radiographic opacification of the paranasal sinuses.

  6. Extravascular eosinophils: Biopsy including artery, arteriole, or venule showing accumulations of eosinophils in extravascular areas.

Definition of ANCA-associated vasculitides in the nomenclature of systemic vasculitis adopted in 2012 by the Chapel Hill consensus conference
Large vessel vasculitis: Giant-cell arteritis; Takayasu arteritis.
Medium-sized-vessel vasculitis: Polyarteritis nodosa; Kawasaki disease.
Small vessel vasculitis*:
  ANCA-associated vasculitides**
   Granulomatosis with polyangiitis (Wegener’s).
   Necrotizing granulomatous inflammation usually involving the upper and lower respiratory tract, and necrotizing vasculitis affecting predominantly small to medium vessels (eg, capillaries, venules, arterioles, arteries, and veins). Necrotizing glomerulonephritis is common.
   Eosinophilic granulomatosis with polyangiitis (Churg-Strauss syndrome).
   Eosinophil-rich and necrotizing granulomatous inflammation often involving the respiratory tract, and necrotizing vasculitis predominantly affecting small to medium vessels, and associated with asthma and eosinophilia. ANCA is more frequent when glomerulonephritis is present.
   Microscopic polyangiitis.
   Necrotizing vasculitis, with few or no immune deposits, predominantly affecting small vessels (ie, capillaries, venules, or arterioles).Necrotizing arteritis involving small and medium arteries may be present. Necrotizing glomerulonephritis is very common. Pulmonary capillaritis often occurs. Granulomatous inflammation is absent.
  Immune complex small-vessel vasculitides
    IgA vasculitis (Henoch-Scho¨nlein purpura)
    Cryoglobulinemic vasculitis
    Hypocomplementemic urticarial vasculitis (anti-C1q vasculitis)
    Antiglomerular basement membrane (antiglomerular basement membrane) disease
Variable vessel vasculitis: Behcet’s disease; Cogan’s syndrome
Single-organ vasculitis: Cutaneous leukocytoclastic angiitis; cutaneous arteritis; primary central nervous system vasculitis; isolated aortitis; others
Vasculitis associated with systemic disease: Lupus vasculitis; rheumatoid vasculitis; sarcoid vasculitis; others
Vasculitis associated with probable etiology: Hepatitis C virus–associated cryoglobulinemic vasculitis; hepatitis B virus–associated vasculitis; syphilis-associated aortitis; drug-associated immune complex vasculitis; drug-associated ANCA-associated vasculitis; cancer-associated vasculitis; others

The final version of the revised 2020 Diagnostic and Classification Criteria for Vasculitis, American College of Rheumatology and European League Against Rheumatism) criteria were not been published at the time of this review article (see the text for details).

*Large vessels are the aorta and its major branches and the analogous veins. Medium vessels are the main visceral arteries and veins and their initial branches. Small vessels are intraparenchymal arteries, arterioles, capillaries, venules, and veins.

**Necrotizing vasculitis, with few or no immune deposits, predominantly affecting small vessels (ie, capillaries, venules, arterioles, and small arteries), associated with myeloperoxidase (MPO) ANCA or proteinase 3 (PR3) ANCA. Not all patients have ANCA. Add a prefix indicating ANCA reactivity (eg, MPO-ANCA, PR3-ANCA, ANCA-negative).

Epidemiology

AAV affect men and women equally. The average age at diagnosis is in the fifth or sixth decade, but young children and older adults can be affected.1417 Although AAV have been described in all ethnicities, most patients are Caucasian or Hispanic (93%-98%). The global incidence of AAV between Japan and the United Kingdom is similar, but MPO-AAV is 10 times more frequent than PR3-AAV in Japan.18

The estimated annual incidences of GPA or MPA are close and vary from 2 to 30 cases per million population each, with prevalences of 25-250 cases per million population.19,20 EGPA is much rarer than GPA or MPA, with an incidence of 1-4 cases per million population and a prevalence of about 10-25 cases per million population.21 Intriguingly, the overall incidence rates of AAV increased steadily in the 1980s and 1990s but appear to have stabilized since the early 2000s.14,22,23 The improvements in the recognition of AAV and increased availability of ANCA testing are possible explanations.14,22 Indeed, the increased awareness of physicians has led to shorter diagnostic delay, with some studies in Sweden reporting a median time of only 2 months between the first symptoms and the diagnosis of GPA.23 Finally, a few studies suggested some seasonal variations in the incidence of AAV but not reproducibly during the same periods of the year.19

Genetics

AAV are not considered inherited diseases, and familial occurrence is rare.24 However, several genetic associations have been found with AAV, especially for molecules of the major histocompatibility complex. The strongest associations are with the ANCA antigenic specificity rather than clinical syndrome (MPA vs GPA). PR3-AAV is associated with HLA-DP, the genes encoding alpha1-antitrypsin (SERPINA1) and PR3 (PRTN3), whereas MPO-AAV is associated more often with HLA-DQ. Previous studies already showed an association with more severe forms of GPA and allele deficiency of alpha-1 antitrypsin (homozygosity for deficiency ZZ, SS, or SZ).25 PTPN22 and the CTLA4 locus appear to have a role in AAV susceptibility.2628 SEMA6A has been associated with an increased risk of GPA, but more studies are needed to validate this association.26,28 A study demonstrated a 73-fold increased frequency of HLA-DRB1*15 alleles in African Americans with PR3-AAV, a minority of the AAV population.29

HLA-DRB4 gene, present in carriers of HLADRB1*04, HLA-DRB1*07, or HLA-DRB1*09 alleles, is a genetic risk factor for the development of vasculitic manifestations of EGPA, whereas HLA-DRB3 and HLA-DRB1*13 are associated with decreased likelihood.25 Also, the HLA-DQ region is associated with MPO-ANCA–positive but not ANCA–negative EGPA,30 which reinforces the idea of two subtypes of EGPA, as discussed later in this article.

Clinical and Biological Findings, Diagnosis

Classification but not diagnostic criteria are available for AAV. Updated classification criteria from the Diagnostic and Classification Criteria for Vasculitis, American College of Rheumatology, and European League Against Rheumatism have been recently developed and should be published in early 2022, after having been fully endorsed. The previous classification criteria and the nomenclature definitions of AAV are in Table 1.

The main characteristics and differences for GPA, MPA, and EGPA are summarized in Table 2, and Figures 1 to 8 show some of the most typical manifestations. AAV are all potentially life-threatening, but limited and/or less severe forms exist. The limited form of GPA has slightly different definitions according to different study groups but is defined, as in the WGET trial, as GPA without life- or organ-threatening manifestations and without gastrointestinal, ocular, or central nervous system involvement.31 GPA localized to the upper airway is often persistent and refractory to treatment, with frequent relapses. The main target organs of GPA are the upper and lower respiratory tracts, lungs, and kidneys. MPA is a nongranulomatous vasculitis, and mostly affects the lungs and kidneys, like GPA but without the formation of nodules. EGPA’s hallmark is lateonset asthma and eosinophilia, with other vasculitic manifestations.3234 Other than the most common disease manifestations listed in Table 2, some rarer include parotid gland involvement, retroperitoneal fibrosis, pancreatitis, splenic infarct, and genitourinary lesions, such as prostatitis, orchitis, and penile necrosis.

Table 2.

Main characteristics of the three ANCA-associated vasculitides.

Microscopic polyangiitis Eosinophilic granulomatosis with polyangiitis (Churg - Strauss syndrome) Granulomatosis with polyangiitis
Clinical manifestations
  Constitutional symptoms (fever, arthralgia, myalgia) 55%-80% 30%-50% 70 %-100%
  Skin Purpura (35-60%) Purpura, pseudourticarial rash (50-70%) Purpura (10-50%)
  ENT manifestations Few patients (2-30%), not specific, not destructive, and not granulomatous Frequent (20-80%): Allergic rhinitis, sinus polyposis (not destructive) Frequent (50-95%): Crusting rhinitis, destructive sinusitis, saddle-nose deformity, nasal septum deformity, otitis media, decreased/loss of smell or taste, gum hypertrophy/ pain
  Lung involvement Frequent (60-80%): Alveolar hemorrhage Frequent (50%): transient patchy infiltrates, eosinophil pleural effusion, rarely nodules, alveolar hemorrhage (rare; 3-10%) Frequent (60-80%): lung solid and/or excavated nodules, alveolar hemorrhage, bronchial and/or subglottic stenosis
  Asthma No Yes (100%) No
  Kidney involvement Very frequent: Glomerulonephritis (necrotizing extracapillary), 80% Not frequent: Glomerulonephritis (necrotizing extracapillary), 20% Frequent: Glomerulonephritis (necrotizing extra-capillary): 60-80 %
  Peripheral neuropathy (mononeuritis multiplex) Possible (35%) Very frequent (65-75%) Possible (25%)
  Other “classical’’ manifestations Venous thrombosis 7-8% Cardiac manifestations (1050%; cardiomyopathy); venous thrombosis 7-8% Eye manifestations ( scleritis, orbital tumor); pachymeningitis; venous thrombosis 7-8 %
Biology
  Standard Nonspecific inflammatory syndrome; check creatinine and urine analysis (red blood cell casts?) Eosinophilia, often >3,000/ mm3Nonspecific inflammatory syndrome Nonspecific inflammatory syndrome; check creatinine and urine analysis ( red blood cell casts?)
  ANCA Yes (60-80%): mainly MPOANCA, peri-nuclear-ANCA (p-ANCA) Yes (30-40%): mainly MPOANCA, peri-nuclear-ANCA (p-ANCA) Yes (90% if systemic disease): mainly PR3-ANCA, cytoplasmic-ANCA ( c-ANCA )
Radiology Check chest X-ray and/or CT scan for alveolar hemorrhage (ground-glass opacities); other imaging studies according to clinical presentation Check chest X-ray and/or CT scan for labile and transient lung infiltrates (rarely alveolar hemorrhage or nodules); sinus X-ray and/or CT scan for nonerosive sinusitis, polyps; other imaging studies according to clinical presentation Check chest X-ray and/or CT scan for alveolar hemorrhage (ground-glass opacities), lung nodules, excavated or not, subglottic and/or bronchial stenosis; sinus X-ray and/or CT scan for erosive sinusitis, pseudotumor; other imaging according to clinical presentation
Histology Necrotizing vasculitis of small-sized vessels; no granuloma (rare cases) Granuloma, including eosinophils (frequent); necrotizing vasculitis of small-sized vessels Granuloma (frequent but not always); necrotizing vasculitis of small-sized vessels

ANCA, antineutrophil cytoplasm antibody; CT, computed tomography; ENT, ear - nose - throat; MPO, myeloperoxidase; PR3, proteinase 3.

Figure 1.

Figure 1.

Saddle-nose deformity in a patient with granulomatosis with polyangiitis ( GPA ).

Figure 8.

Figure 8.

Histology of muscle-nerve biopsy in a patient with EGPA. Note the massive vessel wall and peri-vascular infiltrate by inflammatory cells, mainly eosinophils, the vessel wall necrosis and subsequent vessel lumen occlusion.

Besides these classical and well-known manifestations of AAV, an association between interstitial lung disease (ILD) and AAV has been established over the last few years. This complication is mostly seen in patients over 65 years of age, Japanese patients, and/or those with MPO-ANCA–positive disease (70 % of cases of AAV-associated ILD). The prevalence of ILD is reported in 23% of patients with GPA and up to 45% of patients with MPA.35 Lung fibrosis can precede the development of AAV, as seen in almost half of the reported patients, by a few months to 12 years.35,36 On imaging, patterns encountered are usual interstitial pneumonia in 50%71% of cases, nonspecific interstitial pneumonia in 7%-31%, and desquamative interstitial pneumoniae in up to 14%.35,37 The presence of ILD implies poor prognosis, with increased mortality and a median survival of 5 years,35 , 36 but more studies are needed to confirm this point and determine the best treatment approach for these patients.

Patients with AAV are also at increased risk of venous thromboembolism, with an estimated frequency of 8%. Venous thrombosis is reported especially when the disease is active, just prior to, then within the first 6 months after a disease flare (diagnosis or relapse) but can also occur, at lesser frequency, during phases of remission.38 , 39

Several studies showed an increased likelihood of cardiovascular events (CVEs) in patients with AAV, mostly because of accelerated atherosclerosis, and these events usually occur in the first 5 years after diagnosis.40 , 41 Patients have a threefold higher risk of CVEs than the general population, and the risk is more than eightfold increased with cerebrovascular accidents.41 The incidence rates of CVEs were 19 per 1000 patient-years in a Canadian study and 27.8 per 1000 patient-years in France.42 , 43

The diagnosis of AAV relies on the combination of clinical findings and results of imaging studies, basic and nonspecific biology tests (inflammatory markers such as C-reactive protein level, complete blood count, renal parameters, and urine sediment analysis), and more specific ones, including ANCA testing of course and, when feasible, a biopsy of an affected organ. For quantitative detection of ANCA, the recommendation is to use highquality antigen-specific immunoassays such as enzyme-linked immunosorbent assay (ELISA) or chemiluminescent immunoassay as the preferred first screening test.44 Many centers have almost abandoned ANCA testing by indirect immunofluorescence (to detect c-ANCA with a cytoplasmic labeling pattern, p-ANCA with a perinuclear pattern, and sometimes x-ANCA for an atypical pattern). In all, 60%-80% of patients with MPA are ANCApositive, mostly MPO-ANCA, whereas 90% of patients with severe GPA are ANCA-positive, mostly PR3-ANCA.31 Patients with ANCAnegative GPA most often have limited disease, but the disease can progress to a more severe form and sometimes become ANCA-positive. Approximately 30%-40% of EGPA patients are ANCA-positive, mostly MPO-ANCA.32,45

Ruling out the differentials, including infections or malignancy, is mandatory when suspecting the diagnosis or a relapse, as well as complications of therapy for the latter. Of note, a positive ANCA test result can be observed in other conditions, such as auto-immune hepatitis, ulcerative colitis, infection with hepatitis C virus or HIV, or infectious endocarditis, without associated vasculitis. In the latter condition, the specificity of ANCA is often different from PR3or MPO-ANCA.44 Some drugs can induce AAVs (propylthiouracil is the most famous one), usually with high titers of MPO-ANCA.44 , 46 Levamisole-cocaine use can trigger a vasculopathy with high titers of ANCA (PR3- and/or MPO-ANCA, sometimes with antielastase specificity, that can be detected by nonroutine ELISA). The vasculopathy closely resembles GPA and occasionally requires a similar treatment approach, at least initially, along with cocaine discontinuation.47 Finally, all patients with renal and/or alveolar hemorrhage should also be tested for antiglomerular basement membrane antibody disease, which can mimic AAVs or be an additional condition. ( “Double ANCA and antiglomerular basement membrane positivity” patients have worse renal prognosis and require a slightly different treatment approach.)

Biopsies of skin lesions, present in up to 40%60% of patients with AAVs, are easy to perform and will usually reveal vasculitis, although most medium- or small-sized vessel vasculitis can cause the same type of histological lesions.48 Vascular and extravascular eosinophilic granulomas are more suggestive of EGPA. Nasal or sinus biopsies have low sensitivity (20%-50%) even when performed by trained surgeons on ulcerated areas and with deep mucosa samples. Open lung biopsy yields high sensitivity (80%-90%) but is invasive. The diagnostic value of transbronchial biopsy is only about 10%.49 In patients with renal involvement, renal biopsy can show the hallmark features of pauci-immune necrotizing and crescentic glomerulonephritis and can further be classified according to pathologic activity. Focal glomerular lesions (50% of normal glomeruli) have the best prognosis and usually do not progress to end-stage renal disease (ESRD), whereas sclerotic glomerular lesions (50% sclerotic glomeruli) have the worst prognosis and high mortality rates. Crescentic and mixed categories have an intermediate risk of progression to ESRD.50 On initial presentation, the degree of chronic damage on kidney biopsy seems associated with overall survival and is one of the best predictors of renal outcome.51 In practice, the need for an invasive biopsy in an ANCApositive patient with typical clinical manifestations of AAV and no evidence of infection, cancer, or drug-induced disease remains a case-based decision.

Pathogenesis

The pathogenesis of AAV is a complex process of immune dysregulations with genetic and environmental influences, but the exact cause is not fully elucidated.

The hypothesis of an infectious agent, such as Staphylococcus aureus for GPA, which would (over) activate the immune system, has been repeatedly suggested. However, the infection can barely be sufficient to cause or explain the full-blown disease by itself and its multiple facets.52

The pathogenic role of MPO-ANCA has been supported by animal models by passive transfer of MPO-ANCAs and a single case of MPA in the newborn of a mother with anti-MPO antibodies (passive transplacental transfer).53,54 Additional experiments demonstrated the major importance of neutrophils, the neutrophil activation pathway, and the alternative complement pathway (mainly through C5a) in the MPO-ANCA-induced animal model.55,56 Recent studies showed that blockade of the C5a receptor (CD88) with CCX168 (now known as avacopan) could prevent but also limit renal disease in this murine model.57

Conversely, we lack convincing evidence to support the pathogenic role of PR3-ANCA or to explain why some patients with biopsyconfirmed GPA or MPA have typical clinical manifestations despite being ANCA-negative. There may be several explanations. First, the homology between human and murine PR3 is less than that for MPO. Hence, animal models are more difficult to generate, and complex alterations in mice are required before achieving some vasculitic changes close to those seen in GPA.58,59 Second, only a fraction of ANCAs are pathogenic in an individual (ie, only those ANCAs directed toward one or a few specific epitopes). A study of MPOANCA - positive patients demonstrated only a subset of MPO-ANCAs associated more strongly and specifically with disease activity than others directed toward different MPO epitopes.60 Moreover, these specific ANCAs, directed toward the linear amino-acid sequence 447-459 of the MPO molecule were detected in many ANCA-negative patients with MPA, were well associated with disease activity and, when injected in mice, triggered the development of (proliferative, but not necrotizing) glomerulonephritis. Routine ANCA tests do not properly detect these specific MPO-epitope-ANCAs447-459 because such tests are based on total serum. Serum immunoglobulins (Ig) first need to be purified for detection of these specific MPO-epitopeANCAs447-459. A serum factor (likely a fraction of ceruloplasmin) indeed bound to these specific MPO-epitope-ANCAs447-459, thereby preventing detection of the most clinically relevant ANCAs with routine tests. Specific alterations and “maturation” of ANCAs may also be necessary for them to become pathogenic, including the selection of higheraffinity PR3-ANCAs in nasal mucosa granulomas or modulation of their sialylation levels. 61

An excessive and abnormally sustained antigenic presentation of PR3 and/or MPO has also been implicated (in patients with GPA or MPA) through their overexpression on neutrophils, genetically determined, as well as endothelial cell membranes and the formation by neutrophils of neutrophil extracellular traps and microparticles, which include the PR3 and/or MPO molecules. 52,62

B cells likely play a major role in the pathogenesis, not only because they produce ANCAs. Studies have reported elevated serum levels of B-lymphocyte stimulator (BLys) in patients with AAV, with good association with disease activity. After anti-B-cell treatment with rituximab, serum BLys level is increased in patients with AAVs.63 Imbalances in the different T-cell subtypes (T helper 1 [Th1], Th2, Th17, regulatory CD4þ CD25þ FoxP3þ T cells, etc) and/or cytokine - chemokine networks can also lead to, or at least participate in, rupture of tolerance, triggering autoimmunity and/or an oxidative burst aggressive toward endothelial cells.

The pathogenesis of EGPA is less studied than that of GPA and MPA. Only 30%-40% of patients with EGPA are positive for ANCA, mainly perinuclear MPO-ANCAs.21 We lack an animal model of EGPA, and MPO-ANCA induced murine models do not show any of the main features of EGPA (ie, blood eosinophilia, tissue eosinophilic granulomas, or obstructive lung disease). Eosinophils likely play a central and/or additional role in the development of EGPA, directly or through their granule degradation products. Th2 lymphocytes are also a key part of the disease because they produce specific cytokines (interleukin 4 [IL-4], IL-5, and IL-13). In particular, IL-5 is known to play a key role in the maturation process, activation, proliferation, and survival of eosinophils and is associated with disease activity.64 IL-25, with an increased level in EGPA patients, is secreted by activated eosinophils and enhances Th2 cytokine production, thus promoting an inflammatory cycle.65 IL-10 level is also increased in EGPA, mostly in ANCA-negative patients, via gene polymorphisms, and promotes the dysregulated Th2 pathway and increases IgG4 level.65 Finally, the efficacy of B-cell depletion with rituximab, at least in some patients with EGPA, has questioned the contribution of B lymphocytes to the pathogenesis of EGPA. Patients with relapsing EGPA show an elevated number of CD80þ, CD27þ, and CD95þ B cells and lower percentages of CD19þ cells.66

Treatment Approaches

Current standard of care for induction therapy in GPA and MPA

The current treatment of severe AAV involves two phases: Remission induction therapy based on the combination of glucocorticoids and another immunosuppressive agent, then once remission is achieved, maintenance therapy (to maintain remission). New treatment regimens have been developed and studied over the past three decades to limit the toxicity of agents that have been used as conventional therapies for many years, such as cyclophosphamide and glucocorticoids.9 Table 3 summarizes the indications and adverse events associated with the most commonly used treatments.67

Table 3.

Main current medications for the treatment of ANCA-associated vasculitides: Principles of use and main risks. See text for details and latest published guidelines or recommendations.

Medication Current validated indications Dosage Adverse events
Glucocorticoids Cornerstone treatment for remission induction Induction: Oral prednisone (or prednisolone-equivalent) 1 mg/kg/ day Infections, diabetes, hypertension, osteoporosis, gastritis, peptic ulcers, weight gain, avascular necrosis, myopathy, neuropsychiatric manifestations, cataracts, skin thinning, cardiovascular disease, fluid retention.
Continuous low-dose glucocorticoids might have an additive role as maintenance therapy Often preceded by IV methylprednisolone pulses (7.5-15 mg/kg/day, up to 1000 mg, daily for 1-3 days)
Cyclophosphamide Remission induction therapy in patients with severe AAVs Oral: 2 mg/kg/day (maximum 200 mg/day) or Intravenous: 15 mg/kg (maximum 1200 mg) every 2 weeks for the first three doses, then every 3 weeks for the next 3-6 doses Infections (patients must receive prophylactic therapy against Pneumocystis jiroveci pneumonia), myelosuppression and cytopenias, nausea, vomiting, myocarditis, hemorrhagic cystitis (mesna to consider for bladder protection with IV cyclophosphamide), infertility, primary ovarian failure, teratogenicity.
Dose to adjust according to age and renal function (fixed-500 mg per pulse can be considered in older patients) - Increased risk of malignancy, such as skin cancers, acute leukemia, myeloid malignancies, bladder cancer.
Rituximab Remission induction therapy in patients with severe AAVs Induction: 375 mg/m2 IV weekly for four doses or 1000 mg IV at days 1 and 15 Infections, including hepatitis B virus reactivation (screening for hepatitis B virus prior to treatment) or P. jiroveci pneumonia (patients must receive prophylactic therapy), infusion reactions, arthralgias, skin rash, hypogammaglobulinemia ( IgG, M and A levels to monitor in patients with serious or recurrent infections), late-onset neutropenia.
Maintenance of remission in patients with severe AAVs Maintenance: 500-1000 mg IV every 4-6 months (500 mg every 6 months most commonly used) for at least 18 months (four doses)
Methotrexate Remission induction therapy in patients with limited AAVs and/or without life-threatening disease 0.3 mg/kg/week (oral or subcutaneous) Maximum dose of 25 mg/week Infections, oral ulcers (folic acid supplementation to limit this risk), transaminitis and hepatotoxicity, gastrointestinal symptoms (nausea/vomiting, diarrhea, abdominal pain), skin rashes, alopecia, headache, fatigue, myelosuppression, pneumonitis, pericarditis, teratogenicity, impairment of fertility.
Maintenance of remission Avoid in renal impairment (glomerular filtration rate <30 mL/min) - Can increase the risk of neoplasia, such as lymphoproliferative disorders.
Azathioprine Maintenance of remission 2 mg/kg/day (oral) Maximum 200 mg/day Cytopenias (patients with homozygous deficiency of thiopurine methyltransferase at high risk), infection, hepatotoxicity, pancreatitis, skin rashes, nausea and vomiting, diarrhea, alopecia.
-Can increase the risk of neoplasia, such as lymphoproliferative disorders and nonmelanoma skin cancers.
-Safe in pregnancy.
Mycophenolate mofetil Remission induction therapy in patients with limited AAVs and/or without life-threatening disease 2 g/day (oral) Maximum dose of 3 g/day Infections, gastrointestinal symptoms (diarrhea, abdominal pain, nausea/vomiting), cytopenias, teratogenicity.
Maintenance of remission in patients with contraindications or adverse events to azathioprine or methotrexate - Can increase the risk of neoplasia, such as lymphoproliferative disorders and nonmelanoma skin cancers.
Mepolizumab Refractory and/or relapsing nonsevere EGPA and/or glucocorticoiddependent patients (prednisone 7.5 mg/day) Subcutaneous injection of 300 mg every 4 weeks Headache, nasopharyngitis, sinusitis, upper respiratory tract infections, arthralgias (including back pain) and myalgias, abdominal pain, infusion reactions, pruritis and eczema, shingles.

ANCA, antineutrophil cytoplasm antibody; EGPA, eosinophilic granulomatosis with polyangiitis; IV, Intravenous.

Remission is usually achieved by a combination of high doses of glucocorticoids with cyclophosphamide or rituximab for 36 months.68 The optimal dose, tapering regimen, and duration of glucocorticoid treatment are still uncertain. For severe organ involvement, such as alveolar hemorrhage or rapidly progressive glomerulonephritis, patients typically receive intravenous pulses of methylprednisolone (500-1000 mg for 13 days), although strong evidence for this practice is lacking and the treatment seems associated with severe infection.69 Prednisone is typically given afterward at 1 mg/kg/day (not exceeding 80 mg/day) and progressively tapered after 2 weeks, by about 10% every 2 weeks. Hence, in most studies, the prednisone dose was tapered to 5-10 mg/day at 46 months of induction therapy. The randomized multicentric trial PEXIVAS, recently published, studied a reduced-dose regimen of glucocorticoids (prednisone tapered from 1 to 0.5 mg/kg/day after only 1 week, then weekly or biweekly, until reaching 5 mg/day at the end of month 4). The regimen was found noninferior to the standard regimen in patients with severe AAV in terms of deaths and ESRD. At 6 months, the cumulative dose of oral glucocorticoids in the reduced-dose group was less than 60% of that in the standard-dose group and was associated with a reduced rate of severe infections.70

Cyclophosphamide and rituximab are the two possible agents for remission induction, combined with glucocorticoids, in patients with severe GPA or MPA. The drugs can induce remission in more than 80% of patients. Cyclophosphamide was first used in the mid-1970s, and its efficacy is well proven. The CYCAZAREM trial showed that oral cyclophosphamide (2 mg/kg/day) combined with glucocorticoids for 3-6 months induced remission in 93% of patients.7 Subsequently, two trials showed that intravenous pulsed (see Table 3 for dosing regimen) or oral daily cyclophosphamide were as effective in inducing remission.71,72 However, long-term follow-up showed more relapses with intravenous cyclophosphamide but no differences between the administrations in renal function or survival.72 Conversely, intravenous pulses were associated with less cumulative cyclophosphamide dose exposure, thereby less risk of infertility, late cancer (mainly bladder cancer or lymphoma), and leukopenia.71 Cyclophosphamide toxicity can be minimized by reducing the dose in older patients.73

Given the associated risk with cumulative use of cyclophosphamide, rituximab was studied as an alternative therapy in the early 2000s. The RAVE trial enrolled patients with newly diagnosed or relapsing severe GPA or MPA and showed that rituximab (375 mg/m2 weekly for four doses) was noninferior to cyclophosphamide in inducing remission. The trial found a trend toward superiority of rituximab for relapsing disease and, in a posthoc subgroup analysis, for patients with PR3ANCA–positive AAV.10,74 Whereas there was no difference in terms of infection rates between the arms in the RAVE trial, some subsequent real-life data showed that RTX-based induction therapy was also associated with less serious infectious complications, and was less risky to use than cyclophosphamide in patients with known, concurrent infections.75,76 The RITUXVAS trial randomized patients with severe, newly diagnosed AAV with renal involvement to receive glucocorticoids and rituximab plus two intravenous pulses of cyclophosphamide (experimental arm) or the standard intravenous cyclophosphamide pulse therapy for 3-6 months. Both groups achieved similar sustained remission rates at 12 months.12 Rituximab should clearly be preferred in patients with contraindications and/or at risk of infertility with cyclophosphamide. A different dosing regimen of rituximab (1 g on days 1 and 15) has also been used for induction. The regimen is more practical, and a recent meta-analysis revealed equivalency to the 375-mg/m2-weekly-for-4-weeks regimen, as used in the RAVE and RITUXVAS trials.77 To help minimize glucocorticoid exposure (and cyclophosphamide toxicity), two small observational studies studied a combination of rituximab and cyclophosphamide, not so different from that used in the experimental arm of RITUXVAS. More than 80% of patients achieved remission at 6 months, with fewer cumulative doses of glucocorticoids.78,79 Clinical controlled trials are needed to confirm these findings and better delineate the place, if any, of the combined use of cyclophosphamide and rituximab, which may carry an increased risk of infections.

The additive role of plasma exchange has been debated for years for severe AAV, especially for patients with renal involvement and/ or alveolar hemorrhage. In the MEPEX trial in the early 2000s, plasma exchange reduced the risk of progression to ESRD by 24% at month 12 (P ¼ .03), but the long-term follow-up (4 years) revealed no difference in mortality and ESRD.80,81 The factorial-designed PEXIVAS trial, mentioned above, enrolled 704 patients with severe AAV, most with glomerulonephritis and some with diffuse alveolar hemorrhage. Patients were randomly assigned to undergo plasma exchange (7 plasma exchanges within 14 days) or no plasma exchange. The study did not find any reduced incidence of ESRD or death at up to 7 years of follow-up, in the entire study population or in any subgroup analyses, although the latter analyses were underpowered.70 The results of a recent cohort analysis of more than 400 patients with severe renal disease further support the results of PEXIVAS.82

Mycophenolate mofetil (MMF) and methotrexate combined with glucocorticoids can be considered for remission induction in patients with nonsevere GPA or renal but nonsevere MPA. Methotrexate was compared to cyclophosphamide for remission induction in nonsevere, nonrenal GPA or MPA, and remission at 6 months was similar in both groups. However, methotrexate was associated with a longer time to remission in patients with more severe disease and with a higher number of relapses at 18 months.83 MMF was compared to intravenous cyclophosphamide for remission induction in a randomized controlled trial in newly diagnosed GPA or MPA without life-threatening disease and was also found noninferior at 6 months. However, at 18 months, high relapse rates also occurred in the MMF group, mostly in PR3-ANCA–positive patients.84 Abatacept (CTLA4-Ig) is currently being studied as another possible therapeutic option for relapsing and nonsevere GPA (ABROGATE; ClinicalTrials.gov Identifier: NCT02108860) based on the promising results of a small open-label series.

Glucocorticoids may also be used alone, as first-line therapy, to induce remission in the rare patients with nonsevere, nonrenal MPA (ie, with a five-factor score of 085) However, more than half of these patients eventually require the addition of another immunosuppressant because of progressive, refractory or relapsing disease. The CHUSPAN2 study showed no benefit of a combination of glucocorticoids and azathioprine (vs glucocorticoids and a placebo of azathioprine) as first-line treatment for these rare patients.86 In France, a study is evaluating a combination of glucocorticoids and rituximab (vs glucocorticoids and a placebo of rituximab) in these patient populations (RITUXGOPRO; ClinicalTrials.gov Identifier: NCT03920722).

Forthcoming, probable changes in induction treatment of GPA and MPA

As emphasized in the pathogenesis section, the alternative complement pathway is important in AAV. A few years ago, a small open-label study of avacopan, the oral selective C5a receptor inhibitor, showed that it could be noninferior to prednisone as a remission-induction treatment in AAV, combined with cyclophosphamide or rituximab.87 The larger, randomized, and double-blinded ADVOCATE trial then evaluated avacopan (30 mg twice daily for 1 year) versus prednisone (1 mg/kg/day initially, then tapered and stopped at month 6) as additional therapy to cyclophosphamide or rituximab in AAV.88 The results of this study showed that 72% of patients achieved disease remission at 26 weeks in the avacopan group (and no glucocorticoids) as compared with 70% in the standard-treatment group with glucocorticoids (noninferior, but not significantly better). This response was sustained at 52 weeks, with disease remission rates of 66% and 55% (P < .01), respectively. Subgroup analyses also suggested even better results with avacopan in the patients treated with rituximab for induction, those MPO-ANCA - positive and/or relapsers. The proportion of serious adverse events was comparable with avacopan and glucocorticoids (37% and 39%), and improvement in renal function was significantly greater with avacopan at both 26 and 52 weeks.88 The use of avacopan instead of (or with much less) glucocorticoids may be the next “revolution” in AAV treatment since the discovery of the efficacy of rituximab. However, there may still be someplace for highdose glucocorticoids, at least initially, in very severe and/or refractory disease, or maybe at a lower dose (5 mg per day) for maintenance, as detailed below. The optimal duration of avacopan also needs to be established and its long-term safety determined.

Current standard of care for maintenance therapy in GPA and MPA

Following induction therapy with glucocorticoids and cyclophosphamide or rituximab, 7090% of patients will achieve remission. However, continued treatment with an immunosuppressant is required to prevent disease relapse.89 Cyclophosphamide is not continued beyond the time of remission as maintenance therapy because of its significant toxicity. A switch to azathioprine was found equally effective at preventing relapses, occurring at about 14% at 1-year postremission.7 In the WEGENT trial, azathioprine and methotrexate were compared as maintenance agents, following cyclophosphamide-based induction, and showed similar relapse rates and proportion of adverse events.6 Conversely, MMF was found less effective than azathioprine for maintaining remission in the IMPROVE trial, but it can still be considered in patients intolerant to all the other maintenance options.8 Leflunomide can also be used, but the evidence is more limited. Rituximab has now been studied in many cohorts and trials for maintenance therapy in AAV and was found the most efficient agent to date at preventing relapses. Hence, many groups now recommend rituximab as the first choice for maintenance in AAV. In investigating maintenance of remission, following induction with glucocorticoids and cyclophosphamide, the French MAINRITSAN trial showed the superiority of rituximab (500 mg at remission, again 15 days later, then every 6 months for a total of 2 years) at 28 months as compared with azathioprine in patients with newly diagnosed or relapsing GPA or MPA.13 The extended followup at 60 months confirmed the clear and sustained superiority of rituximab, even after it had been stopped (major relapse-free survival rates at 60 months of 72 vs 49%; P ¼ .003).90 Subsequent studies showed that the second maintenance dose (500 mg at day 15) could be omitted. The more recently completed RITAZAREM trial followed 170 patients with relapsing disease who received induction therapy with rituximab and glucocorticoids. Patients were then maintained with rituximab (1000 mg every 4 months, for 2 years) or azathioprine. Rituximab was again found superior to azathioprine in preventing disease relapse (18% vs 38% of major relapses at 24 months; P < .001).91 The optimal regimen needed for rituximab infusions for maintenance may still need refinements and likely patient-based adjustments. Most patients may receive 500 mg every 6 months once remission has been achieved; some may need 1000 mg every 4 months, perhaps those with more severe, relapsing disease. The MAINRITSAN-2 trial studied another approach: A group of patients received 500 mg every 6 months, and the other group received 500 mg at the time of remission but then repeat infusions only when CD19þ B lymphocytes reappeared or with markedly increased ANCA titers (reappearance or twofold increase) based on measurements every 3 months. The relapse rates did not differ between the two groups, and the “tailored-arm” group received fewer infusions.92 This tailored regimen could be an alternative maintenance regimen in some patients, either immediately after the remission has been achieved or after the 2-year mark of maintenance with systematic rituximab infusions. It is associated with lower infusion costs but requires close clinical and biological monitoring.

Other treatments have been studied for the maintenance of remission. Sulfamethoxazole/ trimethoprim (800 mg/160 mg twice daily; cotrimoxazole) can be used as an adjunct maintenance treatment in patients with limited GPA, but its efficacy at preventing relapse is debated and not retained in recent metaanalyses.93,94 Etanercept has not been found effective and was associated with an increased risk of malignancy.95 Belimumab is a human monoclonal antibody against BLyS that was studied in combination with azathioprine for maintenance of remission in newly diagnosed or relapsing severe AAVs. The trial results did not show a reduction in relapse risk as compared with controls receiving placebo and azathioprine. However, patients who achieved remission after induction with rituximab (rather than cyclophosphamide) and who subsequently received belimumab did not experience any disease relapses.96 A clinical trial is ongoing to compare rituximab monotherapy to a sequential combination of rituximab and belimumab for remission maintenance in patients with PR3-ANCA–positive disease (COMBIVAS; ClinicalTrials.gov Identifier: NCT03967925).

The optimal duration of maintenance therapy should be at least 24 months.68 Past the 24month mark, the continuation or not of maintenance therapy is, in practice, often individualized according to several patient characteristics, including the ANCA serotype (PR3-ANCA - positive patients relapse more), persistence of ANCAs after induction therapy (associated with a higher risk of relapse as well), previous relapse history (associated with higher risk for further relapses), organ involvement and/or the patient’s or physician’s preference. The REMAIN trial showed that prolonged use of azathioprine and low-dose prednisone beyond 24 months was associated with a further reduction in relapse risk and improved renal survival at 48 months after diagnosis.97 The MAINRITSAN-3 randomized placebo-controlled trial enrolled 97 patients with AAVs who had achieved remission after 18 months of rituximab maintenance therapy in MAINRITSAN-2 trial. Patients then received rituximab 500 mg or placebo every 6 months for an additional 18 months. Rituximab continuation beyond 2 years was again associated with lower relapse rates than with placebo, with no increase in adverse event rates. At 56 months, relapse-free survival rates were 96% versus 74% in the rituximab and placebo groups (P ¼ .008).98 Although this difference was statistically significant, it is debatable to treat all patients for 4 years, when threequarters of them will remain disease-free if rituximab is stopped at year 2. As mentioned above, the longer treatment may be preferable for PR3-ANCA - positive patients with relapsing disease until an alternative even more effective than rituximab is available.

Treatment of EGPA

Treatment of EGPA also consists of a remission induction therapy followed by a maintenance phase, but there are several nuances and differences as compared with that for GPA and MPA. An internal consensus task force for EGPA established that most patients with nonsevere forms of the disease can initially be treated with glucocorticoids alone.99 The French CHUSPAN trial showed that in 72 patients with newly diagnosed EGPA without poor prognosis factors, a treatment solely with glucocorticoids achieved remission in 93% of cases. However, 35% of the patients showed relapse, mostly upon glucocorticoid tapering and in the first year of treatment, which required the addition of another immunosuppressant.100 The addition of azathioprine to glucocorticoids for such patients, tested as first-line treatment, versus glucocorticoids alone in the CHUSPAN2 trial did not result in lower relapse rates, lower exacerbation rates of asthma or ear-nose-throat manifestations or any significant glucocorticoid sparing.86 Other agents may achieve better results, but only rituximab (still combined with glucocorticoids) is being investigated as possible first-line therapy in patients with nonsevere EGPA (REOVAS; ClinicalTrials.gov Identifier: NCT02807103; results expected late 2021 or early 2022).

Patients with life- and/or organ-threatening EGPA should unequivocally receive glucocorticoids and an additional immunosuppressant (cyclophosphamide) for 3-6 months.99 A study comparing 6 or 12 intravenous cyclophosphamide pulses in patients with EGPA showed six pulses associated with more relapses (86% vs 62%; P ¼ .07). However, patients were not receiving subsequent maintenance therapy, and, eventually, a large proportion, 74%, exhibited clinical relapse.101 Thus, maintenance therapy is needed, and usually azathioprine or methotrexate is used.99 Leflunomide or MMF can also be used, but data are lacking to clearly recommend one agent over another for remission maintenance in EGPA.45 A study is comparing azathioprine to rituximab for maintenance therapy in patients with EGPA (MAINRITSEG; ClinicalTrials.gov Identifier: NCT03164473; recruiting). The optimal duration of treatment is definitely not known in EGPA, but the consensus is that it should be given for at least 18-24 months following remission induction.68 The duration can also be adjusted based on individual patient characteristics. Patients with ANCA-positive EGPA showed a higher risk of relapse despite better survival rates than ANCA-negative patients.32 The latter patients have more frequent cardiac manifestations and lung infiltrates, whereas patients with ANCA-positive EGPA have more frequent purpura, mononeuritis multiplex, lung hemorrhage, and renal manifestations.32

Glucocorticoids should be gradually tapered until withdrawal, when possible, but many EGPA patients require long-term prednisone because of steroid-dependent asthma and/or ear - nose - throat manifestations. In a French cohort, 84% of patients required ongoing glucocorticoids.32

The evidence for rituximab as part of the induction therapy in EGPA is limited, definitively not yet as strong as for GPA and MPA. Rituximab can be considered in selected patients, especially ANCA-positive patients with refractory disease or renal involvement.99 In one study of 41 patients with EGPA, mainly with refractory or relapsing disease, rituximab achieved remission in 49% by 12 months; ANCA-positive patients achieved remission more frequently. However, only 6% of patients completely tapered their glucocorticoids at 12 months.102 Another recent study showed similar results in 69 patients, but rates of asthma or sinonasal relapses remained frequent.103 Case reports have also described severe bronchospasms after rituximab infusions, so caution is warranted in EGPA. The French REOVAS and MAINRITSEG studies, mentioned earlier, will help determine the place of rituximab for EGPA induction and/or maintenance.

Mepolizumab, an anti-IL-5 humanized monoclonal antibody, was recently studied in a randomized placebo-controlled study. In this MIRRA study, 136 patients with relapsing or refractory EGPA received monthly subcutaneous injections of 300 mg mepolizumab over 52 weeks versus placebo. Importantly, the trial excluded patients with the newly diagnosed disease and/or active severe disease. Remission, as defined in the protocol, lasting more than 24 cumulative weeks, was achieved in 28% of patients in the mepolizumab group versus 3% with placebo. Glucocorticoids had been discontinued at month 12 in (only) 18% of mepolizumab patients as compared with 3% of placebo patients.104 Hence, clinical benefit was significantly higher for patients receiving mepolizumab than placebo, but 47% of the former patients still failed to achieve remission, and 56% showed relapse. Patients with higher eosinophil count had a better response to mepolizumab.105 The glucocorticoid-sparing effect of mepolizumab, especially for asthma or other ear-nose-throat manifestations, was further supported by smaller case series.106 Some data recently published suggested that mepolizumab at only 100 mg monthly, instead of 300 mg, could achieve some improvement, although to a slightly lesser extent and with some concern about a rebound of disease if control is not optimal with this lower dosage.107,108 More studies of mepolizumab in EGPA are needed and of other anti-IL-5 agents, including benralizumab (MANDARA; ClinicalTrials.gov Identifier: NCT04157348) or reslizumab (ClinicalTrials.gov Identifier: NCT02947945).

Immunoglobulins can be considered secondline therapy for EGPA flares refractory to other treatments, especially in pregnancy or, based mostly on case reports, in the context of myocardial or nerve involvement.99 Omalizumab, a humanized anti-IgE monoclonal antibody, has some proven efficacy in allergic asthma and rhinitis. A study of 17 patients with EGPA and severe steroid-dependent asthma showed that omalizumab had a steroid-sparing effect, but the medication was discontinued during follow-up because of refractory disease in 25% of patients, with relapse in half of them.109 Another recent study showed similar results in 18 other patients, with mild improvement of asthma and/or sinonasal symptoms, some glucocorticoid-sparing effect, but a significant number of patients showed relapse after the tapering of glucocorticoids.110 The risk of severe EGPA flares after reduction of glucocorticoids raises the question of the safety of omalizumab, and its benefit in EGPA is limited.

Follow-up, Prognosis, and Disease Assessment

As mentioned above, with current optimal therapies, the rate of remission at 6 months in all AAV is 80%-90%.90,111 Patients with major organ involvement have poorer prognosis, and mortality rate remains around 10%, especially during the first year of treatment, because of the underlying vasculitis, or its treatment-related side effects, mainly infections.112,113 Relapses remain frequent in GPA (up to 30% at 4 years from diagnosis, when using maintenance rituximab for 2 years), a little bit less in MPA (around 10% at 4 years, from diagnosis, when using maintenance rituximab for 2 years) and EGPA (up to 35 % ). However, in EGPA, up to 80% of the patients have persistent and/or “relapsing” asthma or sinus problems, thus remain steroid-dependent.32,114,115

Diagnosing and treating promptly relapses can be challenging, as there are no good predictive or diagnostic markers. Serial measurement of ANCA is by default still the “best” available predictive option but lacks both specificity and sensitivity.116 Following ANCA (and CD19þ B cells) in patients treated with rituximab may have better predictive value than with conventional immunosuppressive drugs such as azathioprine.74,92,117 When a relapse is suspected, it is important to rule out mimickers, especially (opportunistic) infections or, more rarely, other complications of treatments, such as hematuria due to cyclophosphamide-related bladder cancer, or new lung nodules related to malignancy.

A few disease assessment tools exist, such as the Birmingham Vasculitis Activity Score (BVAS, version 3) or the BVAS/WG ( Wegener’s granulomatosis).118120 Because they have been developed to assess disease activity in clinical trials, they have limited interest in routine practice, other than reminding physicians of all the main organ manifestations to check in patients with AAV. Damage can also be assessed in trials using scores such as the Vasculitis Damage Index. Studies in AAV showed that no more than 10%-20% of patients will keep a VDI ¼ 0, which supports the need to identify treatment strategies with fewer side effects and more rapidly effective.121,122

Conclusion

Management of AAV has evolved tremendously in the past two decades, with substantial improvements in survival and quality of life. Induction treatment is now well codified, but some major changes may again occur soon. Our expanding knowledge of the pathogenesis and genetic contribution has provided new therapeutic targets, such as the C5a receptor antagonist (avacopan) for GPA and MPA. The optimal duration of low-dose glucocorticoids therapy and the most effective regimen for rituximab-based maintenance therapy are still under study. Reducing the need for or, at least, the cumulative use of glucocorticoids may become a reality with some of these newly developed treatments. The identification of reliable biological markers remains needed to better assess disease activity, predict disease relapse, and further personalize the treatment approach.

A few patients still experience refractory disease and/or unrelenting relapses, which underscores the continuing need for newer and more effective therapies. Treatment of multirelapsing or refractory disease, with or without complex manifestations, such as subglottic and tracheobronchial stenoses, orbital tumor, or pachymeningitis, goes beyond the scope of this review article. Such diseases should ideally be managed in reference centers with expertise in vasculitis.

Many other questions of importance remaining to be answered include how to reduce the damage associated with the disease (ESRD, peripheral nerve damage, saddle-nose deformity in GPA) or its treatments and how best to treat associated nonvasculitic manifestations, such as asthma in EGPA. Finally, because all these new treatments and/or biologics reviewed in this article are expensive, cost-effectiveness studies are needed. Biosimilars of these agents are not as expensive, and their preferential use over the originator is now mandated in many countries, but data on their safety and efficacy in AAV remain very limited.123,124

Figure 2.

Figure 2.

Nasal septum perforation in a patient with GPA. Note the light going from one nostril to the other through the perforated nasal septum and the crusty bleeding posterior wall of the sinonasal cavity.

Figure 3.

Figure 3.

Purpuro-ecchymotic skin lesions on the legs of a patient with eosinophilic granulomatosis with polyangiitis (EGPA). Legs are the most involved areas for skin lesions in antineutrophil cytoplasm antibody (ANCA)associated vasculitides.

Figure 4.

Figure 4.

Nodular cutaneous lesions in a patient with GPA. Elbows are a frequent location for such skin lesions in GPA and EGPA.

Figure 5.

Figure 5.

Computed tomography (CT) scan of sinuses in a patient with GPA. Note the perforated nasal septum and bilateral opacification of maxillary sinuses - sinusitis.

Figure 6.

Figure 6.

Chest CT of a patient with GPA. Note the multiple solid lung nodules, surrounded by ground-glass opacities, suggestive of associated peri-nodular alveolar hemorrhage.

Figure 7.

Figure 7.

Chest CT in a patient with microscopic polyangiitis (MPA). Note the diffuse ground-glass opacities, suggestive of alveolar hemorrhage, with some pseudonodular consolidation appearance in the left lung.

Funding Statement

The authors declared that this study has eceived no financial support.

Footnotes

Peer-review: Externally peer-reviewed.

Author Contributions: Concept - CP; Design - CR, JPM, CP; Supervision - CP; Materials - CP; Literature Search - CR, JPM, CP; Writing Manuscript - CR, JPM, CP; Critical Review - CR, JPM, CP.

Declaration of Interests: Dr. Pagnoux reports receiving fees for serving on advisory boards from Roche, Sanofi, ChemoCentryx, AstraZeneca, and GlaxoSmithKline; lecture fees from Roche, GlaxoSmithKline; and educational grant support from Roche, GlaxoSmithKline, and Amgen. Dr. Ross and Makhzoum have no conflict of interest to report.

Funding: Dr. Pagnoux reports receiving fees for serving on advisory boards from Roche, Sanofi, ChemoCentryx, AstraZeneca, and GlaxoSmithKline; lecture fees from Roche, GlaxoSmithKline; and educational grant support from Roche, GlaxoSmithKline, and Amgen. Drs. Ross and Makhzoum have no financial disclosures to report.

References

  • 1.Jennette JC, Falk RJ, Bacon PA, et al. Revised international chapel hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013;65(1):1–11. [DOI] [PubMed] [Google Scholar]
  • 2.Masi AT, Hunder GG, Lie JT, et al. The American College of Rheumatology 1990 criteria for the classification of Churg-Strauss syndrome (allergic granulomatosis and angiitis). Arthritis Rheum. 2010;33(8):1094–1100. [DOI] [PubMed] [Google Scholar]
  • 3.Leavitt RY, Fauci AS, Bloch DA, et al. The American College of Rheumatology 1990 criteria for the classification of Wegener’s granulomatosis. Arthritis Rheum. 2010;33(8):1101–1107. [DOI] [PubMed] [Google Scholar]
  • 4.Jennette JC, Falk RJ, Andrassy K, et al. Nomenclature of systemic vasculitides. Proposal of an international consensus conference. Arthritis Rheum. 1994;37(2):187–192. [DOI] [PubMed] [Google Scholar]
  • 5.Craven A, Robson J, Ponte C, et al. ACR/EULARendorsed study to develop diagnostic and classification criteria for vasculitis (DCVAS). Clin Exp Nephrol. 2013;17(5):619–621. [DOI] [PubMed] [Google Scholar]
  • 6.Pagnoux C, Mahr A, Hamidou MA, et al. Azathioprine or methotrexate maintenance for ANCA-associated vasculitis. N Engl J Med. 2008;359(26):2790–2803. [DOI] [PubMed] [Google Scholar]
  • 7.Jayne D, Rasmussen N, Andrassy K, et al. A randomized trial of maintenance therapy for vasculitis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med. 2003;349(1):36–44. [DOI] [PubMed] [Google Scholar]
  • 8.Hiemstra TF, Walsh M, Mahr A, et al. Mycophenolate mofetil vs azathioprine for remission maintenance in antineutrophil cytoplasmic antibody-associated vasculitis: A randomized controlled trial. JAMA. 2010;304(21):2381–2388. [DOI] [PubMed] [Google Scholar]
  • 9.Walsh M, Merkel PA, Peh CA, et al. Plasma exchange and glucocorticoid dosing in the treatment of anti-neutrophil cytoplasm antibody associated vasculitis (PEXIVAS): Protocol for a randomized controlled trial. Trials. 2013;14:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stone JH, Merkel PA, Spiera R, et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. N Engl J Med. 2010;363(3):221–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Specks U, Merkel PA, Seo P, et al. Efficacy of remission-induction regimens for ANCAassociated vasculitis. N Engl J Med. 2013;369(5):417–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jones RB, Tervaert JW, Hauser T, et al. Rituximab versus cyclophosphamide in ANCAassociated renal vasculitis. N Engl J Med. 2010;363(3):211–220. [DOI] [PubMed] [Google Scholar]
  • 13.Guillevin L, Pagnoux C, Karras A, et al. Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis. N Engl J Med. 2014;371(19):1771–1780. [DOI] [PubMed] [Google Scholar]
  • 14.Mohammad AJ. An update on the epidemiology of ANCA-associated vasculitis. Rheumatology (Oxford). 2020;59(Supplement_3):iii42–iii50. [DOI] [PubMed] [Google Scholar]
  • 15.Pearce FA, Grainge MJ, Lanyon PC, Watts RA, Hubbard RB. The incidence, prevalence and mortality of granulomatosis with polyangiitis in the UK Clinical Practice Research Datalink. Rheumatology (Oxford). 2016;56(4):589–596. [DOI] [PubMed] [Google Scholar]
  • 16.Pearce FA, Lanyon PC, Grainge MJ, et al. Incidence of ANCA-associated vasculitis in a UK mixed ethnicity population. Rheumatology (Oxford). 2016;55(9):1656–1663. [DOI] [PubMed] [Google Scholar]
  • 17.Watts RA, Mahr A, Mohammad AJ, Gatenby P, Basu N, Flores-Suarez LF. Classification, epidemiology and clinical subgrouping of antineutrophil cytoplasmic antibody (ANCA)associated vasculitis. Nephrol Dial Transplant. 2015;30(Suppl 1):i14–i22. [DOI] [PubMed] [Google Scholar]
  • 18.Fujimoto S, Watts RA, Kobayashi S, et al. Comparison of the epidemiology of anti-neutrophil cytoplasmic antibody-associated vasculitis between Japan and the U.K. Rheumatology (Oxford). 2011;50(10):1916–1920. [DOI] [PubMed] [Google Scholar]
  • 19.Mohammad AJ, Jacobsson LT, Westman KW, Sturfelt G, Segelmark M. Incidence and survival rates in Wegener’s granulomatosis, microscopic polyangiitis Churg-Strauss syndrome and polyarteritis nodosa. Rheumatology (Oxford). 2009;48(12):1560–1565. [DOI] [PubMed] [Google Scholar]
  • 20.Hellmich B, Lamprecht P, Spearpoint P, et al. New insights into the epidemiology of ANCAassociated vasculitides in Germany: Results from a claims data study. Rheumatology (Oxford). 2021;60:4868–4873. [DOI] [PubMed] [Google Scholar]
  • 21.Pagnoux C. Churg-Strauss syndrome: Evolving concepts. Discov Med. 2010;9(46):243252. [PubMed] [Google Scholar]
  • 22.Berti A, Cornec D, Crowson CS, Specks U, Matteson EL. The epidemiology of antineutrophil cytoplasmic autoantibody-associated vasculitis in Olmsted county, Minnesota: A twenty-year US population-based study. Arthritis Rheumatol. 2017;69(12):2338–2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Nelveg-Kristensen KE, Szpirt W, Carlson N, et al. Increasing incidence and improved survival in ANCA-associated vasculitis—A Danish nationwide study. Nephrol Dial Transplant. 2020. DOI: 10.1093 /ndt/gfaa 303. [DOI] [PubMed]
  • 24.Knight A, Sandin S, Askling J. Risks and relative risks of Wegener’s granulomatosis among close relatives of patients with the disease. Arthritis Rheum. 2008;58(1):302–307. [DOI] [PubMed] [Google Scholar]
  • 25.Wieczorek S, Holle JU, Epplen JT. Recent progress in the genetics of Wegener’s granulomatosis and Churg-Strauss syndrome. Curr Opin Rheumatol. 2010;22(1):8–14. [DOI] [PubMed] [Google Scholar]
  • 26.Xie G, Roshandel D, Sherva R, et al. Association of granulomatosis with polyangiitis (Wegener’s) with HLA-DPB1*04 and SEMA6A gene variants: Evidence from genome-wide analysis. Arthritis Rheum. 2013;65(9):2457–2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lyons PA, Rayner TF, Trivedi S, et al. Genetically distinct subsets within ANCA-associated vasculitis. N Engl J Med. 2012;367(3):214–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rahmattulla C, Mooyaart AL, van Hooven D, et al. Genetic variants in ANCA-associated vasculitis: A meta-analysis. Ann Rheum Dis. 2016;75(9):1687–1692. [DOI] [PubMed] [Google Scholar]
  • 29.Cao Y, Schmitz JL, Yang J, et al. DRB1*15 allele is a risk factor for PR3-ANCA disease in African Americans. J Am Soc Nephrol. 2011;22(6):11611167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lyons PA, Peters JE, Alberici F, et al. Genomewide association study of eosinophilic granulomatosis with polyangiitis reveals genomic loci stratified by ANCA status. Nat Commun. 2019;10(1):5120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Stone JH. Limited versus severe Wegener’s granulomatosis: Baseline data on patients in the Wegener’s granulomatosis etanercept trial. Arthritis Rheum. 2003;48(8):2299–2309. [DOI] [PubMed] [Google Scholar]
  • 32.Comarmond C, Pagnoux C, Khellaf M, et al. Eosinophilic granulomatosis with polyangiitis (Churg-Strauss): Clinical characteristics and long-term followup of the 383 patients enrolled in the French Vasculitis Study Group cohort. Arthritis Rheum. 2013;65(1):270–281. [DOI] [PubMed] [Google Scholar]
  • 33.Sinico RA, Bottero P. Churg-Strauss angiitis. Best Pract Res Clin Rheumatol. 2009;23(3):355366. [DOI] [PubMed] [Google Scholar]
  • 34.Keogh KA, Specks U. Churg-Strauss syndrome: Clinical presentation, antineutrophil cytoplasmic antibodies, and leukotriene receptor antagonists. Am J Med. 2003;115(4):284–290. [DOI] [PubMed] [Google Scholar]
  • 35.Alba MA, Flores-Suarez LF, Henderson AG, et al. Interstital lung disease in ANCA vasculitis. Autoimmun Rev. 2017;16(7):722–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Katsumata Y, Kawaguchi Y, Yamanaka H. Interstitial lung disease with ANCA-associated vasculitis. Clin Med Insights Circ Respir Pulm Med. 2015;9(Suppl 1):51–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sacoto G, Boukhlal S, Specks U, et al. Lung involvement in ANCA-associated vasculitis. La Presse Med. 2020;49(3):104039. [DOI] [PubMed] [Google Scholar]
  • 38.Novikov P, Makarov E, Moiseev S, et al. Venous thromboembolic events in systemic vasculitis. Ann Rheum Dis. 2015;74(3):e27. [DOI] [PubMed] [Google Scholar]
  • 39.Allenbach Y, Seror R, Pagnoux C, et al. High frequency of venous thromboembolic events in Churg-Strauss syndrome, Wegener’s granulomatosis and microscopic polyangiitis but not polyarteritis nodosa: A systematic retrospective study on 1130 patients. Ann Rheum Dis. 2009;68(4):564–567. [DOI] [PubMed] [Google Scholar]
  • 40.Houben E, Mendel A, van der Heijden JW, et al. Prevalence and management of cardiovascular risk factors in ANCA-associated vasculitis. Rheumatology (Oxford). 2019;58(12):2333–2335. [DOI] [PubMed] [Google Scholar]
  • 41.Berti A, Matteson EL, Crowson CS, Specks U, Cornec D. Risk of cardiovascular disease and venous thromboembolism among patients with incident ANCA-associated vasculitis: A 20year population-based cohort study. Mayo Clin Proc. 2018;93(5):597–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Avina-Zubieta JA, Mai A, Amiri N, et al. Risk of myocardial infarction and stroke in patients with granulomatosis with polyangiitis (Wegener’s): A population-based study. Arthritis Rheumatol. 2016;68(11):2752–2759. [DOI] [PubMed] [Google Scholar]
  • 43.Mourguet M, Chauveau D, Faguer S, et al. Increased ischemic stroke, acute coronary artery disease and mortality in patients with granulomatosis with polyangiitis and microscopic polyangiitis. J Autoimmun. 2019;96:134141. [DOI] [PubMed] [Google Scholar]
  • 44.Bossuyt X, Cohen Tervaert JW, Arimura Y, et al. Position paper: Revised 2017 international consensus on testing of ANCAs in granulomatosis with polyangiitis and microscopic polyangiitis. Nat Rev Rheumatol. 2017;13(11):683–692. [DOI] [PubMed] [Google Scholar]
  • 45.Ennis D, Lee JK, Pagnoux C. Mepolizumab for the treatment of eosinophilic granulomatosis with polyangiitis. Expert Opin Biol Ther. 2019;19(7):617–630. [DOI] [PubMed] [Google Scholar]
  • 46.Moiseev S, Cohen Tervaert JW, Arimura Y, et al. 2020 international consensus on ANCA testing beyond systemic vasculitis. Autoimmun Rev. 2020;19(9):102618. [DOI] [PubMed] [Google Scholar]
  • 47.Pearson T, Bremmer M, Cohen J, Driscoll M. Vasculopathy related to cocaine adulterated with levamisole: A review of the literature. Dermatol Online J. 2012;18(7):1. [PubMed] [Google Scholar]
  • 48.Marzano AV, Raimondo MG, Berti E, Meroni PL, Ingegnoli F. Cutaneous manifestations of ANCA-associated small vessels vasculitis. Clin Rev Allergy Immunol. 2017;53(3):428–438. [DOI] [PubMed] [Google Scholar]
  • 49.Masiak A, Zdrojewski Z, Peksa R, et al. The usefulness of histopathological examinations of non-renal biopsies in the diagnosis of granulomatosis with polyangiitis. Reumatologia. 2017;5(5):230–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Berden AE, Ferrario F, Hagen EC, et al. Histopathologic classification of ANCA-associated glomerulonephritis. J Am Soc Nephrol. 2010;21(10):1628–1636. [DOI] [PubMed] [Google Scholar]
  • 51.Berti A, Cornec-Le Gall E, Cornec D, et al. Incidence, prevalence, mortality and chronic renal damage of anti-neutrophil cytoplasmic antibody-associated glomerulonephritis in a 20-year population-based cohort. Nephrol Dial Transplant. 2019;34(9):1508–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jennette JC, Falk RJ, Gasim AH. Pathogenesis of antineutrophil cytoplasmic autoantibody vasculitis. Curr Opin Nephrol Hypertens. 2011;20(3):263–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bansal PJ, Tobin MC. Neonatal microscopic polyangiitis secondary to transfer of maternal myeloperoxidase-antineutrophil cytoplasmic antibody resulting in neonatal pulmonary hemorrhage and renal involvement. Ann Allergy Asthma Immunol. 2004;93(4):398–401. [DOI] [PubMed] [Google Scholar]
  • 54.Xiao H, Heeringa P, Hu P, et al. Antineutrophil cytoplasmic autoantibodies specific for myeloperoxidase cause glomerulonephritis and vasculitis in mice. J Clin Invest. 2002;110(7):955963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Xiao H, Schreiber A, Heeringa P, Falk RJ, Jennette JC. Alternative complement pathway in the pathogenesis of disease mediated by antineutrophil cytoplasmic autoantibodies. Am J Pathol. 2007;170(1):52–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Xiao H, Heeringa P, Liu Z, et al. The role of neutrophils in the induction of glomerulonephritis by anti-myeloperoxidase antibodies. Am J Pathol. 2005;167(1):39–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Xiao H, Dairaghi DJ, Powers JP, Ertl LS, et al. C5a receptor (CD88) blockade protects against MPO-ANCA GN. J Am Soc Nephrol. 2014;25(2):225–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Little MA, Al-Ani B, Ren S, et al. Anti-proteinase 3 anti-neutrophil cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a humanized immune system. PLoS One. 2012;7(1):e28626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.van Timmeren MM, Heeringa P. Pathogenesis of ANCA-associated vasculitis: Recent insights from animal models. Curr Opin Rheumatol. 2012;24(1):8–14. [DOI] [PubMed] [Google Scholar]
  • 60.Roth AJ, Ooi JD, Hess JJ, et al. Epitope specificity determines pathogenicity and detectability in ANCA-associated vasculitis. J Clin Invest. 2013;123(4):1773–1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Espy C, Morelle W, Kavian N, et al. Sialylation levels of anti-proteinase 3 antibodies are associated with the activity of granulomatosis with polyangiitis (Wegener’s). Arthritis Rheum. 2011;63(7):2105–2115. [DOI] [PubMed] [Google Scholar]
  • 62.Kessenbrock K, Krumbholz M, Schonermarck U, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009;15(6):623625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Prendecki M, Pusey CD. Recent advances in understanding of the pathogenesis of ANCAassociated vasculitis. F1000Res. 2018;7:1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Trivioli G, Terrier B, Vaglio A. Eosinophilic granulomatosis with polyangiitis: Understanding the disease and its management. Rheumatology (Oxford). 2020;59(Suppl 3): iii84-iii 94. [DOI] [PubMed]
  • 65.Wu EY, Hernandez ML, Jennette JC, Falk RJ. Eosinophilic granulomatosis with polyangiitis: Clinical pathology conference and review. J Allergy Clin Immunol Pract. 2018;6(5):1496–1504. [DOI] [PubMed] [Google Scholar]
  • 66.Tsurikisawa N, Saito H, Oshikata C, Tsuburai T, Akiyama K. Decreases in the numbers of peripheral blood regulatory T cells, and increases in the levels of memory and activated B cells, in patients with active eosinophilic granulomatosis and polyangiitis. J Clin Immunol. 2013;33(5):965–976. [DOI] [PubMed] [Google Scholar]
  • 67.Mendel A, Ennis D, Go E, et al. CanVasc consensus recommendations for the management of antineutrophil cytoplasm antibody-associated vasculitis: 2020 update. J Rheumatol. 2021;48:555–566. [DOI] [PubMed] [Google Scholar]
  • 68.Yates M, Watts RA, Bajema IM, Cid MC, et al. EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis. Ann Rheum Dis. 2016;75(9):1583–1594. [DOI] [PubMed] [Google Scholar]
  • 69.Chanouzas D, McGregor JAG, Nightingale P, et al. Intravenous pulse methylprednisolone for induction of remission in severe ANCA associated vasculitis: A multi-center retrospective cohort study. BMC Nephrol. 2019;20(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Walsh M, Merkel PA, Peh CA, et al. Plasma exchange and glucocorticoids in severe ANCA-associated vasculitis. N Engl J Med. 2020;382(7):622–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.de Groot K, Harper L, Jayne DR, et al. Pulse versus daily oral cyclophosphamide for induction of remission in antineutrophil cytoplasmic antibody-associated vasculitis: A randomized trial. Ann Intern Med. 2009;150(10):670–680. [DOI] [PubMed] [Google Scholar]
  • 72.Harper L, Morgan MD, Walsh M, et al. Pulse versus daily oral cyclophosphamide for induction of remission in ANCA-associated vasculitis: Long-term follow-up. Ann Rheum Dis. 2012;71(6):955–960. [DOI] [PubMed] [Google Scholar]
  • 73.Pagnoux C, Quemeneur T, Ninet J, et al. Treatment of systemic necrotizing vasculitides in patients aged sixty-five years or older: Results of a multicenter, open-label, randomized controlled trial of corticosteroid and cyclophosphamide-based induction therapy. Arthritis Rheumatol. 2015;67(4):1117–1127. [DOI] [PubMed] [Google Scholar]
  • 74.Unizony S, Villarreal M, Miloslavsky EM, et al. Clinical outcomes of treatment of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis based on ANCA type. Ann Rheum Dis. 2016;75(6):1166–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Thomas K, Argyriou E, Kapsala N, et al. Serious infections in ANCA-associated vasculitides in the biologic era: Real-life data from a multicenter cohort of 162 patients. Arthritis Res Ther. 2021;23(1):90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gregersen JW, Chaudhry A, Jayne DR. Rituximab for ANCA-associated vasculitis in the setting of severe infection. Scand J Rheumatol. 2013;42(3):207–210. [DOI] [PubMed] [Google Scholar]
  • 77.Benard V, Farhat C, Zarandi-Nowroozi M, et al. Comparison of two rituximab regimens for induction of remission in antineutrophil cytoplasmic antibody-associated vasculitis: Systemic review and meta-analysis. ACR Open Rheumatol. 2021 Jul;3(7):484-494. [DOI] [PMC free article] [PubMed]
  • 78.McAdoo SP, Medjeral-Thomas N, Gopaluni S, et al. Long-term follow-up of a combined rituximab and cyclophosphamide regimen in renal anti-neutrophil cytoplasm antibodyassociated vasculitis. Nephrol Dial Transplant. 2019;34(1):63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cortazar FB, Muhsin SA, Pendergraft WF III et al. Combination therapy with rituximab and cyclophosphamide for remission induction in ANCA vasculitis. Kidney Int Rep. 2018;3(2):394402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jayne DR, Gaskin G, Rasmussen N, et al. Randomized trial of plasma exchange or highdosage methylprednisolone as adjunctive therapy for severe renal vasculitis. J Am Soc Nephrol. 2007;18(7):2180–2188. [DOI] [PubMed] [Google Scholar]
  • 81.Walsh M, Casian A, Flossmann O, et al. Longterm follow-up of patients with severe ANCAassociated vasculitis comparing plasma exchange to intravenous methylprednisolone treatment is unclear. Kidney Int. 2013;84(2):397402. [DOI] [PubMed] [Google Scholar]
  • 82.Casal Moura M, Irazabal MV, Eirin A, et al. Efficacy of rituximab and plasma exchange in antineutrophil cytoplasmic antibodyassociated vasculitis with severe renal disease. J Am Soc Nephrol. 2020. DOI: 10.1681/ASN.2019111197. [DOI] [PMC free article] [PubMed]
  • 83.de Groot K, Rasmussen N, Bacon PA, et al. Randomized trial of cyclophosphamide versus methotrexate for induction of remission in early systemic antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 2005;52(8):2461–2469. [DOI] [PubMed] [Google Scholar]
  • 84.Jones RB, Hiemstra TF, Ballarin J, et al. Mycophenolate mofetil versus cyclophosphamide for remission induction in ANCA-associated vasculitis: A randomised, non-inferiority trial. Ann Rheum Dis. 2019;78(3):399–405. [DOI] [PubMed] [Google Scholar]
  • 85.Samson M, Puechal X, Devilliers H, et al. Longterm follow-up of a randomized trial on 118 patients with polyarteritis nodosa or microscopic polyangiitis without poor-prognosis factors. Autoimmun Rev. 2014;13(2):197–205. [DOI] [PubMed] [Google Scholar]
  • 86.Puechal X, Pagnoux C, Baron G, et al. Adding azathioprine to Remission-Induction glucocorticoids for eosinophilic granulomatosis with polyangiitis (Churg-Strauss), microscopic polyangiitis, or polyarteritis nodosa without poor prognosis factors: A randomized. Arthritis Rheumatol. 2017;69(11):2175–2186. [DOI] [PubMed] [Google Scholar]
  • 87.Jayne DRW, Bruchfeld AN, Harper L, et al. Randomized trial of C5a receptor inhibitor avacopan in ANCA-associated vasculitis. J Am Soc Nephrol. 2017;28(9):2756–2767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Jayne DRW, Merkel PA, Schall TJ, Bekker P. Avacopan for the treatment of ANCA-associated vasculitis. N Engl J Med. 2021;384(7):599–609. [DOI] [PubMed] [Google Scholar]
  • 89.Jennette JC, Nachman PH. ANCA glomerulonephritis and vasculitis. Clin J Am Soc Nephrol. 2017;12(10):1680–1691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Terrier B, Pagnoux C, Perrodeau E, et al. Longterm efficacy of remission-maintenance regimens for ANCA-associated vasculitides. Ann Rheum Dis. 2018;77(8):1150–1156. [DOI] [PubMed] [Google Scholar]
  • 91.Smith R, Jayne D, Merkel P. A randomized, controlled trial of rituximab versus azathioprine after induction of remission with rituximab for patients with ANCA-associated vasculitis and relapsing disease (abstract). Arthritis Rheumatol. 2019;71 (Suppl 10):1384. https://acrabstracts.org/abstract/a-randomized-controlled-trialof-rituximab-versus-azathioprine-after-induction-of-remission-with-rituximab-for-patientswith-anca-associated-vasculitis-and-relapsingdisease/. Accessed August 24, 2020.
  • 92.Charles P, Terrier B, Perrodeau E, et al. Comparison of individually tailored versus fixedschedule rituximab regimen to maintain ANCA-associated vasculitis remission: Results of a multicentre, randomised controlled, phase III trial (MAINRITSAN2). Ann Rheum Dis. 2018;77(8):1143–1149. [DOI] [PubMed] [Google Scholar]
  • 93.Stegeman CA, Tervaert JW, de Jong PE, Kallenberg CG; Dutch Co-Trimoxazole Wegener Study Group. Trimethoprim-sulfamethoxazole (co-trimoxazole) for the prevention of relapses of Wegener’s granulomatosis. N Engl J Med. 1996;335(1):16–20. [DOI] [PubMed] [Google Scholar]
  • 94.Monti S, Delvino P, Riboli M, et al. The role of trimethoprim/sulfametoxazole in reducing relapses and risk of infections in ANCAassociated vasculitis: A meta-analysis. Rheumatology (Oxford). 2021;60:3553–3564. [DOI] [PubMed] [Google Scholar]
  • 95.Stone JH, Uhlfelder ML, Hellmann DB, Crook S, Bedocs NM, Hoffman GS. Etanercept combined with conventional treatment in Wegener’s granulomatosis: A six-month open-label trial to evaluate safety. Arthritis Rheum. 2001;44(5):1149–1154. [DOI] [PubMed] [Google Scholar]
  • 96.Jayne D, Blockmans D, Luqmani R, et al. Efficacy and safety of belimumab and azathioprine for maintenance of remission in antineutrophil cytoplasmic antibodyassociated vasculitis: A randomized controlled study. Arthritis Rheumatol. 2019;71(6):952–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Karras A, Pagnoux C, Haubitz M, et al. Randomised controlled trial of prolonged treatment in the remission phase of ANCA-associated vasculitis. Ann Rheum Dis. 2017;76(10):16621668. [DOI] [PubMed] [Google Scholar]
  • 98.Charles P, Perrodeau E, Samson M, et al. LongTerm rituximab use to maintain remission of antineutrophil cytoplasmic antibodyassociated vasculitis: A randomized trial. Ann Intern Med. 2020;173(3):179–187. [DOI] [PubMed] [Google Scholar]
  • 99.Groh M, Pagnoux C, Baldini C, et al. Eosinophilic granulomatosis with polyangiitis (ChurgStrauss) (EGPA) consensus task force recommendations for evaluation and management. Eur J Intern Med. 2015;26(7):545–553. [DOI] [PubMed] [Google Scholar]
  • 100.Ribi C, Cohen P, Pagnoux C, et al. Treatment of Churg-Strauss syndrome without poorprognosis factors: A multicenter, prospective, randomized, open-label study of seventy-two patients. Arthritis Rheum. 2008;58(2):586–594. [DOI] [PubMed] [Google Scholar]
  • 101.Cohen P, Pagnoux C, Mahr A, et al. ChurgStrauss syndrome with poor-prognosis factors: A prospective multicenter trial comparing glucocorticoids and six or twelve cyclophosphamide pulses in forty-eight patients. Arthritis Rheum. 2007;57(4):686–693. [DOI] [PubMed] [Google Scholar]
  • 102.Mohammad AJ, Hot A, Arndt F, et al. Rituximab for the treatment of eosinophilic granulomatosis with polyangiitis (Churg-Strauss). Ann Rheum Dis. 2016;75(2):396–401. [DOI] [PubMed] [Google Scholar]
  • 103.Teixeira V, Mohammad AJ, Jones RB, Smith R, Jayne D. Efficacy and safety of rituximab in the treatment of eosinophilic granulomatosis with polyangiitis. RMD Open. 2019;5(1):e000905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wechsler ME, Akuthota P, Jayne D, et al. Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis. N Engl J Med. 2017;376(20):1921–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Steinfeld J, Bradford ES, Brown J, et al. Evaluation of clinical benefit from treatment with mepolizumab for patients with eosinophilic granulomatosis with polyangiitis. J Allergy Clin Immunol. 2019;143(6):2170–2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Moosig F, Gross WL, Herrmann K, Bremer JP, Hellmich B. Targeting interleukin-5 in refractory and relapsing Churg-Strauss syndrome. Ann Intern Med. 2011;155(5):341–343. [DOI] [PubMed] [Google Scholar]
  • 107.Canzian A, Venhoff N, Urban ML, et al. Use of biologics to treat relapsing and/or refractory eosinophilic granulomatosis with polyangiitis: Data from a European Collaborative Study. Arthritis Rheumatol. 2021;73:498–503. [DOI] [PubMed] [Google Scholar]
  • 108.Mukherjee M, Lim HF, Thomas S, et al. Airway autoimmune responses in severe eosinophilic asthma following low-dose Mepolizumab therapy. Allergy Asthma Clin Immunol. 2017;13:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Jachiet M, Samson M, Cottin V, et al. Anti-IgE monoclonal antibody (omalizumab) in refractory and relapsing eosinophilic granulomatosis with polyangiitis (Churg-Strauss): Data on seventeen patients. Arthritis Rheumatol. 2016;68(9):2274–2282. [DOI] [PubMed] [Google Scholar]
  • 110.Celebi Sozener Z, Gorgulu B, Mungan D, et al. Omalizumab in the treatment of eosinophilic granulomatosis with polyangiitis (EGPA): Single-center experience in 18 cases. World Allergy Organ J. 2018;11(1):39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Flossmann O, Berden A, de Groot K, et al. Long-term patient survival in ANCA-associated vasculitis. Ann Rheum Dis. 2011;70(3):488–494. [DOI] [PubMed] [Google Scholar]
  • 112.Wallace ZS, Choi H, Stone JH. Risk of severe infection following rituximab and the efficacy of antimicrobial prophylaxis. Ann Rheum Dis. 2020;79:e40. [DOI] [PubMed] [Google Scholar]
  • 113.Wallace ZS, Lu N, Unizony S, Stone JH, Choi HK. Improved survival in granulomatosis with polyangiitis: A general population-based study. Semin Arthritis Rheum. 2016;45(4):483–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Durel CA, Berthiller J, Caboni S, Jayne D, Ninet J, Hot A. Long-term followup of a multicenter cohort of 101 patients with eosinophilic granulomatosis with polyangiitis (Churg-Strauss). Arthritis Care Res (Hoboken). 2016;68(3):374–387. [DOI] [PubMed] [Google Scholar]
  • 115.Kim MY, Sohn KH, Song WJ, et al. Clinical features and prognostic factors of Churg-Strauss syndrome. Korean J Intern Med. 2014;29(1):8595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Fussner LA, Hummel AM, Schroeder DR, et al. Factors determining the clinical utility of serial measurements of antineutrophil cytoplasmic antibodies targeting proteinase 3. Arthritis Rheumatol. 2016;68(7):1700–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kemna MJ, Damoiseaux J, Austen J, et al. ANCA as a predictor of relapse: Useful in patients with renal involvement but not in patients with nonrenal disease. J Am Soc Nephrol. 2015;26(3):537–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Luqmani RA, Bacon PA, Moots RJ, et al. Birmingham vasculitis activity score (BVAS) in systemic necrotizing vasculitis. QJM. 1994;87(11):671–678. [PubMed] [Google Scholar]
  • 119.Flossmann O, Bacon P, de Groot K, et al. Development of comprehensive disease assessment in systemic vasculitis. Postgrad Med J. 2008;84(989):143–152. [DOI] [PubMed] [Google Scholar]
  • 120.Luqmani RA, Exley AR, Kitas GD, Bacon PA. Disease assessment and management of the vasculitides. Baillieres Clin Rheumatol. 1997;11(2):423–446. [DOI] [PubMed] [Google Scholar]
  • 121.Seo P, Jayne D, Luqmani R, Merkel PA. Assessment of damage in vasculitis: Expert ratings of damage. Rheumatology (Oxford). 2009;48(7):823–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Seo P, Min YI, Holbrook JT, et al. Damage caused by Wegener’s granulomatosis and its treatment: Prospective data from the Wegener’s granulomatosis etanercept trial ( WGET ). Arthritis Rheum. 2005;52(7):2168–2178. [DOI] [PubMed] [Google Scholar]
  • 123.Kwon HC, Kim MK, Song JJ, Park YB, Lee SW. Rituximab biosimilar prevents poor outcomes of microscopic polyangiitis and granulomatosis with polyangiitis as effectively as rituximab originator. Yonsei Med J. 2020;61(8):712–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Mittal S, Naidu G, Sharma A. Response to: Biosimilars for vasculitis: Promise requires validation by quality studies. Clin Rheumatol. 2020;39(10):3155–3156. [DOI] [PubMed] [Google Scholar]

Articles from European Journal of Rheumatology are provided here courtesy of AVES

RESOURCES