Next Article in Journal
Tuberculosis in the Russian Federation: Dynamics of the Epidemic Indicators before and after COVID-19 Pandemic
Previous Article in Journal
Protective Effects of L-2-Oxothiazolidine-4-Carboxylate during Isoproterenol-Induced Myocardial Infarction in Rats: In Vivo Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Type of ANCA May Be Indispensable in Distinguishing Subphenotypes of Different Clinical Entities in ANCA-Associated Vasculitis

by
Afroditi Maria Konstantouli
1,
Georgios Lioulios
1,
Stamatia Stai
1,
Eleni Moysidou
1,
Asimina Fylaktou
2,
Aikaterini Papagianni
1 and
Maria Stangou
1,*
1
Department of Nephrology, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
2
Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Life 2022, 12(10), 1467; https://doi.org/10.3390/life12101467
Submission received: 13 August 2022 / Revised: 15 September 2022 / Accepted: 19 September 2022 / Published: 21 September 2022
(This article belongs to the Section Medical Research)

Abstract

:

Simple Summary

ANCA specificity seems to correlate more accurately with the symptomatology and disease outcome in ANCA-associated vasculitis in comparison to the classification based on clinical syndromes, as described in the Chapel Hill Consensus. This review analyzes the main differences in epidemiology, pathogenesis, histological and clinical manifestations and treatment response according to ANCA type.

Abstract

The traditional nomenclature system for classifying antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) based on clinical phenotype describes granulomatosis with polyangiitis (GPA), eosinophilic granulomatosis with polyangiitis (EGPA) and microscopic polyangiitis (MPA) as distinct clinical entities. This classification has proved its expedience in clinical trials and everyday clinical practice; yet, a substantial overlap in clinical presentation still exists and often causes difficulties in prompt definition and clinical distinction. Additionally, new insights into the AAV pathogenesis point out that PR3 and MPO-AAV may not represent expressions of the same disease spectrum but rather two distinct disorders, as they display significant differences. Thus, it is supported that a classification based on ANCA serotype (PR3-ANCA, MPO-ANCA or ANCA-negative) could be more accurate and also closer to the nature of the disease compared to the phenotype-based one. This review aims to elucidate the major differences between PR3 and MPO-AAV in terms of epidemiology, pathogenesis, histological and clinical manifestations and response to therapeutic approaches.

1. Introduction

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) form a group of autoimmune, systemic, small-vessel vasculitides. Based on the 2012 Revised International Chapel Hill Consensus, the AAV group consists of four clinicopathologic entities: granulomatosis with polyangiitis—GPA (formerly known as Wegener’s granulomatosis), microscopic polyangiitis—MPA, eosinophilic granulomatosis with polyangiitis—EGPA (formerly known as Churg–Strauss syndrome) and renal-limited ANCA-associated vasculitis—RLV [1]. Two main types of ANCA autoantibodies, directed against neutrophil granule components, are detected in the circulation: PR3-ANCA, with specificity for proteinase 3 (PR3), and MPO-ANCA, with specificity for myeloperoxidase (MPO) [2]. Typically, small-sized vessels are involved; glomerular and alveolar capillaries are usually affected, resulting in glomerulonephritis and pulmonary hemorrhage, respectively, followed by skin involvement, manifested as purpuric lesions [3,4]. Other organs, such as the heart, central nervous system and gastrointestinal tract, are less frequently affected. Systemic and general symptoms, such as low-grade fever or malaise, may be the initial manifestations, while multiple tissues are usually involved during disease progression.
Several studies report an important overlap in clinical features between the most common clinical syndromes, GPA and MPA. Therefore, there is a growing assumption that a disease classification based on ANCA serology and not on clinical syndromes could probably differentiate patients much more accurately in terms of symptomatology, outcome and response to different therapeutic approaches. The present review aims to elucidate the major differences between PR3-ANCA and MPO-ANCA regarding epidemiology, pathogenesis, histology, clinical manifestations and treatment outcome and to compare findings with previous classifications based on clinical syndromes.

2. Epidemiology

2.1. Age and Gender

Several studies have validated that the male-to-female ratio is higher in PR3-AAV than in MPO-AAV. A large, multinational cohort showed that the male gender was more common among PR3-ANCA patients (61.5% compared to 43% among MPO-ANCA patients, p < 0.001) [5]. Likewise, a Japanese study reported that the majority of PR3-GPA patients were of male gender, while MPO-GPA is predominated by the female gender [6].
Regarding age, MPO-ANCA patients are usually older, with a median age estimated as 67 years. compared to 60 years., the median age of PR3-ANCA patients [5,6,7].

2.2. Geographic Variation

MPO and PR3-AAV differ significantly in geographic distribution [8] (Figure 1). MPO-ANCA specificity predominates in Asian populations; according to Pearce et al., in Japan, 81.3% of AAV patients were MPO-ANCA(+), whereas PR3-ANCA(+) accounted for only 2.1% of patients [9]. Similarly, a retrospective study of 467 AAV patients in China documented an MPO-ANCA predominance, with a frequency of 91.9% [10]. In contrast, PR3-ANCA positivity is more prevalent in Northern Europe and the Middle East and Turkey (61.2% and 50% of AVV, respectively) [9]. These geographic variations could reflect the effect of different ultraviolet (UV) radiation exposure [5] and distinct genetic backgrounds. The main allele associated with PR3-AAV, HLA-DPB1*04, has a higher frequency in European countries, compared to in Japan and China. Correspondingly, the HLA-DQ variants associated with MPO-AAV are more prevalent in East Asian populations [9].

3. Pathogenesis

3.1. Predisposing Factors

There is a body of growing evidence supporting that a genetically determined background, combined with exposure to environmental factors, can disrupt homeostasis and trigger ANCA production.

3.2. Genetic Associations

Genome-wide association studies have revealed that, although there is no evidence of inheritance in AAV, there is a clear genetic predisposition, associated more strongly with ANCA specificity than with the clinical syndromes [11]. Single-nucleotide polymorphisms (SNPs) in genes of the major histocompatibility complex (MHC) type II have been linked to AAV, with the HLA-DPB1*04 allele being strongly associated with the presence of ANCA [11,12]. PR3-AAV is associated with SNPs at the HLA-DP locus, especially with the HLA-DPB1*04 allele, and MPO-AAV with an SNP at the HLA-DQ locus. The presence of PR3-ANCA is also associated with an SNP at the gene encoding alpha-1-antitrypsin (SERPINA1) and variants of the gene encoding proteinase 3 (PRTN3) [11,12].

3.3. Environmental Factors

3.3.1. Staphylococcus Aureus

It has been proposed that infections could induce loss of tolerance to self-antigens by molecular mimicry and excessive formation of neutrophil extracellular traps (NETs), which lead to prolonged exposure of autoantigens to immune cells [13,14,15]. Although chronic nasal carriage of St. aureus has been documented in MPO-ANCA vasculitis, it is mainly associated with PR3-ANCA patients, increasing their risk of relapse [16,17]. A clinical trial conducted by Salmela et al. found that chronic nasal carriage was almost exclusively a characteristic of PR3-ANCA vasculitis and, when detected in MPO-ANCA patients, was usually intermittent and not chronic [17]. Furthermore, the genetic make-up of St. aureus isolates seems to be different between PR3-ANCA and MPO-ANCA patients, indicating a possibility of distinct pathogenetic mechanisms [18].
Silica: Occupational silica exposure, related to mining, construction work, farming and agriculture, has been recognized as a risk factor for MPO-ANCA vasculitis [8,19]. In a population-based, case-control study, MPO-ANCA specificity was significantly more common than PR3-ANCA specificity among AAV patients with high exposure to silica (67% of MPO-ANCA patients were classified as having high exposure versus 40% of PR3-ANCA patients, p = 0.02) [20].

3.3.2. Latitude and Ultraviolet (UV) Radiation

Epidemiological studies have proved an association between latitude and ANCA serotype. PR3-ANCA-positive vasculitis presents an inverse relation with vitamin-D-effective UV radiation levels. Its incidence increases while moving from south to north, as UV radiation levels decrease [5,21]. The protective role of UV radiation could be attributed to the immunomodulatory effects of vitamin D, which is believed to suppress Th1/Th17 cells and upregulate regulatory T cells [21].

3.3.3. Drug-Induced Vasculitis

Medication is also associated with an ANCA-vasculitis-like syndrome. Drug-induced vasculitis has a milder phenotype, which recedes after discontinuation of the responsible drug [15,22]. MPO-ANCA specificity is more common, but concurrent MPO-ANCA and PR3-ANCA specificity can occur, predominantly in adults, after levamisole-adulterated cocaine inducement [23]. The list of implicated drugs is long and includes propylthiouracil (PTU) and other antithyroid drugs (carbimazole and methimazole), the anti-hypertensive agent hydralazine, the antibiotics minocycline and D-penicillamine and anti-TNFα agents [8,24].

3.4. Immune System Dysfunction

The precise steps triggering loss of self-tolerance to neutrophil antigens remain unknown. However, much attention has been drawn to the potential involvement of a variety of immune system components, including T and B lymphocytes, complement, neutrophil extracellular traps (NETs) and cytokine balance, as explicated below. Pathogenetic mechanisms and involvement of immune dysregulation are described in Figure 2.

3.4.1. T Cells

Increasing evidence suggests an impaired Treg function [15,22,25]. Without the suppressive supervision of Tregs, there is an abnormal expansion of Th17 and effector CD25+ T-cell populations [25]. As a result, elevated levels of T-cell activity markers are detected in AAV patients, e.g., IL-2, IL-17 and IL-23 [22]. These interleukins perpetuate the release of further proinflammatory cytokines, such as TNF-α and IL-1b, that prime neutrophils for ANCA activation [15].

3.4.2. B Cells

Dysregulation of B-cell function at various levels has been described. Wilde et al. showed that AAV patients have diminished levels of IL-10-producing B cells, which regulate T-cell activity (regulatory B cells, Breg) [26]. Breg deficiency leads to insufficient suppression of proinflammatory Th1 cells, promoting autoimmunity. Dysregulated expression of CD19 has also been documented [27]. A subset of memory B cells that express CD19 at higher levels shows loss of tolerance and autoreactivity.

3.4.3. Complement

Stimulated neutrophils release properdin and factor B, positive regulators of the alternate complement pathway, resulting in C5a production [22]. C5a is a powerful chemoattractant that primes further neutrophils for ANCA-induced activation. Thus, the alternative complement pathway comprises an amplification loop [28]. In addition, plasma levels of factor H, a negative regulator of the alternative complement pathway (at the level of C3 convertase), are significantly lower in AAV patients, indicating complement hyperactivation [29].
A prospective study in AAV suggested that activation of the alternative complement pathway has an important role during disease development, but also influences progression [30]. Serum levels of C5a, C3a and Bb were increased and also had a significant correlation with the severity of histologic findings.

3.4.4. NETs

Excessive NET formation, combined with impaired degradation, has a key role in pathogenesis [24,31,32]. NETs constitute an extracellular structure of fibers composed of granule proteins and chromatin. They are released from activated neutrophils and serve antimicrobial purposes by neutralizing a broad spectrum of pathogens [32]. MPO and PR3 antigens are found among the granular components of NETs, and their attachment to chromatin fibers possibly modifies their antigenicity [33]. Furthermore, defective degradation of NETs due to reduced activity of the enzyme DNase I prolongs the exposure of PR3 and MPO antigens and favors their detection by autoreactive, antigen-presenting cells [31,33,34]. ANCAs are thought to perpetuate NET formation by upregulating the production of IL-8, an important cytokine in neutrophil activation and NET generation [34].

3.4.5. Cytokines

The impaired function of innate and adaptive immunity results in elevated amounts of inflammatory mediators, such as IL-6, IL-15, TGF-b1, VEGF, MCP-1 and MIP-1b. Measurement of these cytokines in the serum and urine of AAV patients demonstrated that some of them, such as EGF, IL-2 and IL-9, may have a favorable role, while others, e.g., IL-6, VEGF, IL-15, TGF-β1, MCP-1 and MIP-1β, have a detrimental effect on renal function [35]. Interestingly, there was a discrepancy between PR3 and MPO-AAV in cytokine production. MPO-ANCA patients had increased urinary levels of IL-17, while IL-6, BAFF and IL-5 predominated in the presence of PR3-ANCA.
Likewise, Berti et al. identified distinct profiles of serum circulating cytokines for PR3-AAV and MPO-AAV. PR3-AAV presented significantly higher levels of nine cytokines (IL-6, GM-CSF, IL-15, IL-18, CXCL8/IL-8, CCL-17/TARC, IL-18BP, sIL-2Ra, NGFb), whereas four cytokines were detected at higher levels in MPO-AAV (sIL6R, sTNFRII, NGAL, sICAM-1) [36].

4. Classification

Classifying ANCA-associated vasculitides proves to be a real challenge due to their heterogeneity and significantly overlapping clinical features [37]. One of the most widely used systems is the 2012 Revised Chapel Hill Consensus Conference (CHCC 2012), which provides clear definitions for AAV and highlights the importance of ANCA serology [1]. This nomenclature system categorizes vasculitides based on the size of the affected blood vessels and the clinicopathological findings. AAVs are defined as necrotizing, small-vessel vasculitides (SVV), affecting small intraparenchymal arteries, arterioles, capillaries and venules. The absence of immune deposits (pauci-immune vasculitis) differentiates them from the immune complex SVV group (Figure 3). Furthermore, single-organ AAV (mainly renal-limited, RLV) is introduced as a fourth entity in the group of ANCA-associated vasculitides. The issue of ANCA(−) AAV is also addressed, with it being attributed to an ANCA-independent pathogenesis or to a group of ANCAs undetectable by the currently used methods. Recognizing the prognostic value of ANCAs, the use of a prefix to indicate ANCA serology is advocated, e.g., PR3-GPA, MPO-MPA. Table 1 summarizes the 2012 CHCC definitions for AAV.
For disease activity assessment, the use of the Birmingham Vasculitis Activity Score (BVAS) is advocated. The score is calculated based on signs and symptoms attributable to active vasculitis in nine distinct organ systems [38]. A cross-sectional study for the validation of the new BVAS version (BVAS v.3) indicated a strong correlation between the BVAS score and disease status in AAV patients [39]. The BVAS index has also been shown to have a prognostic value. Patients with a high BVAS score present more relapses and higher mortality [38,40]. Haris et al. documented that PR3-ANCA patients are more likely to have a higher BVAS score and, thus, present a poorer outcome [40].

5. ANCA Biology

The major antigenic targets of ANCA are myeloperoxidase (MPO) and proteinase 3 (PR3), components of cytoplasmic neutrophil granules [41]. MPO-ANCA pathogenicity has been demonstrated in animal models in which the passive transfer of anti-MPO IgG caused necrotizing glomerulonephritis [42]. On the contrary, it has been difficult to confirm the pathogenic role of PR3-ANCA using animal models. Furthermore, there is a discordance between clinical syndromes and ANCA serology. Although GPA is usually associated with PR3-ANCA and MPA with MPO-ANCA, none of the ANCAs is pathognomonic of a clinical syndrome, as MPO-GPA and PR3-MPA can also occur, yet less frequently [41].
Lysosomal membrane protein 2 (LAMP-2), a protein found in neutrophil lysosomes, has also been proposed as an antigenic target of ANCA. In fact, autoantibodies directed against LAMP-2 (anti-LAMP-2) are frequently detected in both MPO and PR3-ANCA-associated glomerulonephritis, as well as in ANCA(−) glomerulonephritis [43,44,45]. New data support the potential pathogenic role of anti-LAMP-2 in renal lesions. Firstly, a membrane glycoprotein of glomerular endothelial cells is structurally related to LAMP-2 and, thus, forms a target of anti-LAMP-2. Furthermore, a sequence homology of LAMP-2 with a bacterial fimbrial protein called FimH is found in many Gram-negative bacteria [46].

6. ANCA as a Prognostic and Follow-Up Biomarker

The utility of ANCA titres for monitoring patients and predicting relapses is still a subject of debate. Thompson et al. reported that patients demonstrating a decrease in PR3-ANCA titres, evaluated by direct ELISA at 4 months after treatment initiation, were more likely to achieve remission in a short period of time [47]. Likewise, Fussner et al. described that an increase in PR3-ANCA levels during complete remission was accompanied by a higher risk of severe relapse within a year, especially among patients with renal involvement or alveolar hemorrhage [48]. Regarding MPO-ANCA, patients with higher titres presented significantly lower renal survival rates at a two-year follow up and were more likely to need permanent dialysis [49]. Thus, monitoring ANCA titres along the course of immunotherapy could help clinicians distinguish patients who are not responding to therapy or suffer a poorer prognosis.

7. Renal Involvement—Histopathology

Renal involvement usually produces pauci-immune, necrotizing glomerulonephritis, with only a few or no immune deposits under immunofluorescence staining [50]. In 2010, an international group of renal pathologists organized the histopathologic findings into four classes of renal lesions based on the proportion between healthy and affected glomeruli [51]:
  • Focal class, if >50% of glomeruli are healthy;
  • Crescentic class, if >50% of glomeruli present crescents;
  • Sclerotic class, if >50% of glomeruli are sclerotic;
  • Mixed class, with no predominance of a lesion phenotype (less than 50% normal, less than 50% crescentic, less than 50% sclerotic glomeruli).
The four distinct glomerular phenotypes respond to the severity of renal damage and have a prognostic value [51,52,53]. The focal class has the best renal survival prognosis, followed by the crescentic and mixed group. On the contrary, the sclerotic phenotype is associated with poor renal outcome.
MPO and PR3-ANCA glomerulonephritis (GN) differ notably in their histopathological presentation. MPO-ANCA GN presents a smaller fraction of healthy glomeruli and more sclerotic glomerular lesions. Interstitial fibrosis and tubular atrophy are also more pronounced in MPO-AAV [54,55]. ANCA(−) vasculitis also shows a higher percentage of sclerotic lesions and interstitial fibrosis, similar to the damages found in MPO-ANCA vasculitis [56]. A recent study that evaluated the association between ANCA specificity and histopathology findings revealed that 64% of PR3-ANCA patients had the crescentic phenotype and only 3% the sclerotic one [57]. On the other hand, renal histology in MPO-ANCA and ANCA(−) patients was similar, with only 28% having the crescentic and 46% the sclerotic phenotype. Table 2 summarizes the main pathogenetic and clinical differences between PR3 and MPO-AAV.

8. Extrarenal Manifestations—ENT, Lung and Ocular Involvement

ENT involvement is usually manifested as chronic granulomatous rhinosinusitis with nasal crusting and epistaxis and less often with septal perforation and saddle nose [58]. Recurrent otitis media and hearing loss can also occur [59]. ENT involvement is strongly associated with PR3-ANCA positivity [60,61]. Similar symptoms can occur in MPO-ANCA patients, but less frequently. A retrospective analysis of three international, multicenter trials showed that ENT involvement, presenting as nasal obstruction and accompanied by granulomatous sinusitis, was definitely more frequent in PR3 compared to in MPO-ANCA patients and also had significant correlation with the degree of renal function impairment and severity of renal histology [60].
Lungs are frequently affected in both PR3 and MPO-AAV. Studies reported that more than 75% of AAV patients have clinical or radiological evidence of lung involvement [62,63]. Central airway disease, in the form of thickening and stenosis, and nodular lesions are the main patterns described in PR3-AAV, whereas interstitial fibrosis, bronchiectasis and alveolar hemorrhage are more prevalent in MPO-AAV [62].
Ophthalmologic disease, such as scleritis and episcleritis, is another common manifestation of AAV and occurs with higher frequency in PR3-AAV than in MPO-AAV. Among the AAV patients with ocular disease followed at the Mayo Clinic from 2003 to 2013, 64% were PR3-ANCA(+), 21% were MPO-ANCA(+) and 15% tested negative for ANCA [64]. The most common symptoms experienced were ocular injection, eye pain and visual acuity loss.
Characteristics of ENT, lung and kidney involvement are depicted in Figure 4.

9. Treatment

The treatment of AAV is based on immunosuppression and involves two phases: the induction phase, lasting between 3 and 6 months, and the maintenance phase once remission is achieved, switching to less toxic immunosuppressants for an additional period of 18–24 months [15,65].
Clinical trials in AAV and their results are summarized in Table 3.

9.1. Induction of Remission

For decades, oral cyclophosphamide combined with glucocorticoids was the first-line treatment for remission induction [66,67,68]. However, the severe, treatment-related toxicity, e.g., leukopenia and life-threatening opportunistic infections, hemorrhagic cystitis, malignancies and infertility, urged the development of safer protocols [69].
The CYCLOPS trial demonstrated that intravenous pulses of cyclophosphamide (15 mg/kg every 2 to 3 weeks) maintained the efficacy of its oral administration at a smaller cumulative dose, producing fewer episodes of severe leukopenia [70]. When long-term relapse data were collected retrospectively, intravenously treated patients had more relapses, but renal function and survival rates were similar in both regimen groups, offering promising results for pulse cyclophosphamide [71]. Interestingly, PR3-ANCA patients were at a higher risk of relapse when treated with cyclophosphamide pulses compared with MPO-ANCA patients, who responded equally to both regimens.
Two randomized trials concluded that rituximab is non-inferior to cyclophosphamide for remission induction. In the RAVE trial, rituximab (375 mg/m2 of body surface per week for 4 weeks) matched the efficacy of oral cyclophosphamide (2 mg/kg per day for 3–6 months, followed by azathioprine) in newly diagnosed disease, including severe renal involvement or alveolar hemorrhage, and was superior in relapsing disease [72]. The induction phase had a duration of 6 months. Regarding ANCA reactivity, PR3-ANCA patients treated with rituximab became ANCA(−) considerably more often than those receiving cyclophosphamide. A subsequent post hoc analysis of the data suggested that rituximab is superior to cyclophosphamide for remission induction in PR3-ANCA patients [73]. Both agents were associated with comparable rates of remission in MPO-ANCA patients.
The RITUXVAS trial compared rituximab with intravenous cyclophosphamide [74]. In this trial, the rituximab-based group also received two pulses of cyclophosphamide due to the relative lack of experience with rituximab at that time. As in the RAVE study, both immunosuppressants demonstrated similar efficacy in remission induction. A RITUXVAS follow-up study showed that mortality, relapses and end-stage renal disease did not differ significantly among patients with rituximab-induced remission without maintenance therapy (apart from with low-dose prednisolone) and cyclophosphamide-induced remission followed by azathioprine [75].
Despite the promising results regarding its efficacy, rituximab yielded similar rates of adverse effects with cyclophosphamide in both trials (RAVE and RITUXVAS), failing to provide any clear safety benefits.

9.2. Maintenance of Remission

A common practice for remission maintenance is converting to azathioprine after cyclophosphamide-induced remission.
The CYCAZAREM trial attempted to determine the optimal time interval for switching to azathioprine [76]. Patients were assigned to two groups, converting to azathioprine after 3–6 or 12 months of cyclophosphamide, respectively. Early cyclophosphamide replacement led to more relapses, but the difference was not found to be statistically important. Additionally, PR3-ANCA patients’ response to treatment did not differ significantly from the other group, although their frequency of relapse was higher than that of MPO-ANCA patients (61% versus 37% in the early replacement group and 41% versus 26% in the second group).
Another question concerned the optimal duration of remission maintenance therapy with azathioprine. The REMAIN study announced that prolonged azathioprine/prednisolone administration, beyond 24 months, increases both relapse-free survival and renal survival [77]. However, patients assigned to the prolonged therapy group were more likely to experience hematological and cardiovascular disorders compared to those who discontinued azathioprine at 24 months. On the contrary, Joode et al. reported that prolongation of azathioprine administration for more than 18 months did not significantly influence the relapse rates, and relapse-free survival depended more on the intensity of induction treatment and ANCA serology (e.g., more relapses in intravenous compared with oral cyclophosphamide and in PR3-ANCA compared with MPO-ANCA patients) [78].
Administration of rituximab for remission maintenance was also evaluated. The MAINRITSAN trial showed that rituximab infusions (500 mg of rituximab on days 0 and 14 and at months 6, 12 and 18, plus daily prednisone) were superior to oral azathioprine (2 mg/kg/day for the first 12 months, followed by 1.5 mg/kg/day for 6 months and 1 mg/kg/day for 4 months, plus daily prednisone) and reduced the frequency of severe relapses (5% in the rituximab group versus 29% in the azathioprine group) [79]. The rate of adverse effects was similar between the two groups.
Given that rituximab failed to reduce the treatment-related toxicity, the key question was whether a smaller cumulative dose remains effective whilst being less toxic. The MAINRITSAN 2 trial compared the standard infusions at fixed intervals (at day 0, day 14 and at month 6, 12 and 18) with tailored rituximab administration based on serological markers (monitoring of C19+ B-cell counts and ANCA levels every 3 months) [80]. Rituximab was reinfused only when one of these biomarkers reappeared or increased considerably. Evaluation at month 28 showed that relapse rates did not differ significantly between the personalized and the fixed-schedule regimen. Additionally, a cohort study conducted at the Mayo Clinic validated the efficacy of rituximab for remission induction and maintenance in PR3-ANCA patients with chronic, refractory disease and documented that reinfusing rituximab after a rise in B-cell count or ANCA titres could prevent an impending relapse [81].

9.3. Considerations before Treatment Selection

PR3-ANCA specificity, granulomatous ENT disease, St. aureus carriage and preserved renal function with low serum creatinine carry a higher relapse risk and might call for a more intense therapeutic strategy [15,67,82]. A higher cumulative dose during induction and prolongation of maintenance treatment are indicated [83].

10. Suggestions for Novel Classification of AAV

As previously defined clinical phenotypes seem not to be able to cover all aspects of AAV, there have been very important efforts to explore disease classification and describe novel subgroups. A remarkable approach was based on cluster analysis, performed in a large cohort of AAV patients. The present analysis started with GPA and MPA diseases and, after the clustering process, which involved multiple correspondence and hierarchical ascendant cluster analysis, resulted in classes with differences in the immune profile and clinical presentation, most significantly in patient outcome [84]. Even more recently, other investigators, again applying unbiased latent class analysis, identified a four-class AAV model, describing a distinct entity affecting young patients presenting with multiple organ involvement and followed by worse outcome [85]. All these attempts simply signify the huge spectrum of AAV, difficulties to define the specific entities and queries regarding treatment approach and outcome but may also indicate the presence of completely divergent diseases not distinctly identified yet.
For the present time, it seems that classification based on ANCA, including PR3-ANCA, MPO-ANCA and ANCA(−), is superior to clinical classification, e.g., GPA, MPA, EGPA and RLD, as it better correlates with disease pathogenesis, histology, clinical symptomatology and outcome.

11. Conclusions

Although the pathogenetic mechanisms of AAV are not yet fully understood, current evidence leads to the conclusion that PR3-ANCA and MPO-ANCA vasculitis differ significantly in prognosis and relapse risk. These differences reflect a need for a therapy tailored to each patient’s specific characteristics. Considering that PR3-ANCA serotype is associated with a higher relapse risk, these patients might need a higher cumulative dose to maintain remission, and, hence, oral cyclophosphamide might be more adequate than its intravenous administration. Furthermore, the data obtained through recent trials indicate that rituximab is a better option for remission induction and maintenance in PR3-ANCA patients. Regarding MPO-ANCA patients, azathioprine seems to be equal to rituximab for remission maintenance. The optimal duration of maintenance treatment remains a controversial issue and could be individualized based on serological biomarkers.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jennette, J.C.; Falk, R.J.; Bacon, P.A.; Basu, N.; Cid, M.C.; Ferrario, F.; Flores-Suarez, L.F.; Gross, W.L.; Guillevin, L.; Hagen, E.C.; et al. 2012 Revised International Chapel Hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013, 65, 1–11. [Google Scholar] [CrossRef]
  2. Morgan, M.D.; Harper, L.; Williams, J.; Savage, C. Anti-neutrophil cytoplasm-associated glomerulonephritis. J. Am. Soc. Nephrol. 2006, 17, 1224–1234. [Google Scholar] [CrossRef]
  3. Flores-Suárez, L.F.; Alba, M.A.; Mateos-Toledo, H.; Ruiz, N. Pulmonary Involvement in Systemic Vasculitis. Curr. Rheumatol. Rep. 2017, 19, 56. [Google Scholar] [CrossRef]
  4. Binda, V.; Moroni, G.; Messa, P. ANCA-associated vasculitis with renal involvement. J. Nephrol. 2018, 31, 197–208. [Google Scholar] [CrossRef]
  5. Weiner, M.; Bjørneklett, R.; Hrusková, Z.; Mackinnon, B.; Poulton, C.J.; Sindelar, L.; Mohammad, A.J.; Eriksson, P.; Gesualdo, L.; Geetha, D.; et al. Proteinase-3 and myeloperoxidase serotype in relation to demographic factors and geographic distribution in anti-neutrophil cytoplasmic antibody-associated glomerulonephritis. Nephrol. Dial. Transplant. 2019, 34, 301–308. [Google Scholar] [CrossRef] [PubMed]
  6. Ono, N.; Niiro, H.; Ueda, A.; Sawabe, T.; Nishizaka, H.; Furugo, I.; Yoshizawa, S.; Yoshizawa, S.; Tsukamoto, H.; Kiyohara, C.; et al. Characteristics of MPO-ANCA-positive granulomatosis with polyangiitis: A retrospective multi-center study in Japan. Rheumatol. Int. 2015, 35, 555–559. [Google Scholar] [CrossRef]
  7. Wójcik, K.; Masiak, A.; Jeleniewicz, R.; Jakuszko, K.; Brzosko, I.; Storoniak, H.; Kur-Zalewska, J.; Wisłowska, M.; Madej, M.; Hawrot-Kawecka, A.; et al. Association of antineutrophil cytoplasmic antibody (ANCA) specificity with the demographic and clinical characteristics of patients with ANCA-associated vasculitides. Pol. Arch. Intern. Med. 2022, 132, 16187. [Google Scholar] [CrossRef]
  8. Scott, J.; Hartnett, J.; Mockler, D.; Little, M.A. Environmental risk factors associated with ANCA associated vasculitis: A systematic mapping review. Autoimmun. Rev. 2020, 19, 102660. [Google Scholar] [CrossRef]
  9. Pearce, F.A.; Craven, A.; Merkel, P.A.; Luqmani, R.A.; Watts, R.A. Global ethnic and geographic differences in the clinical presentations of anti-neutrophil cytoplasm antibody-associated vasculitis. Rheumatology 2017, 56, 1962–1969. [Google Scholar] [CrossRef]
  10. Hong, Y.; Shi, P.; Liu, X.; Yang, L.; Li, K.; Xu, F.; Liang, S.; Liu, Z.; Zhang, H.; Chen, Y.; et al. Distinction between MPO-ANCA and PR3-ANCA-associated glomerulonephritis in Chinese patients: A retrospective single-center study. Clin. Rheumatol. 2019, 38, 1665–1673. [Google Scholar] [CrossRef]
  11. Lyons, P.A.; Rayner, T.F.; Trivedi, S.; Holle, J.U.; Watts, R.A.; Jayne, D.R.; Baslund, B.; Brenchley, P.; Bruchfeld, A.; Chaudhry, A.N.; et al. Genetically Distinct Subsets within ANCA-Associated Vasculitis. N. Engl. J. Med. 2012, 367, 214–223. [Google Scholar] [CrossRef]
  12. Xie, G.; Roshandel, D.; Sherva, R.; Monach, P.A.; Lu, E.Y.; Kung, T.; Carrington, K.; Zhang, S.S.; Pulit, S.L.; Ripke, S.; et al. Association of granulomatosis with polyangiitis (Wegener’s) with HLA-DPB1*04 and SEMA6A gene variants: Evidence grom genome-wide analysis. Arthritis Rheum. 2013, 65, 2457–2468. [Google Scholar] [CrossRef]
  13. Kain, R.; Exner, M.; Brandes, R.; Ziebermayr, R.; Cunningham, D.; Alderson, C.A.; Davidovits, A.; Raab, I.; Jahn, R.; Ashour, O.; et al. Molecular mimicry in pauci-immune focal necrotizing glomerulonephritis. Nat. Med. 2008, 14, 1088–1096. [Google Scholar] [CrossRef]
  14. Geetha, D.; Jefferson, J.A. ANCA-Associated Vasculitis: Core Curriculum 2020. Am. J. Kidney Dis. 2020, 75, 124–137. [Google Scholar] [CrossRef]
  15. Kitching, A.R.; Anders, H.J.; Basu, N.; Brouwer, E.; Gordon, J.; Jayne, D.R.; Kullman, J.; Lyons, P.A.; Merkel, P.A.; Savage, C.O.; et al. ANCA-associated vasculitis. Nat. Rev. Dis. Primers 2020, 6, S267–S276. [Google Scholar] [CrossRef]
  16. Stegeman, C.A.; Cohen Tervaert, J.W.; Sluiter, W.J.; Manson, W.L.; de Jong, P.E.; Kallenberg, C.G.M. Association of Chronic Nasal Carriage of Staphylococcus aureus and Higher Relapse Rates in Wegener Granulomatosis. Ann. Intern. Med. 1994, 120, 12–17. [Google Scholar] [CrossRef]
  17. Salmela, A.; Rasmussen, N.; Tervaert, J.W.C.; Jayne, D.R.W.; Ekstrand, A. Chronic nasal Staphylococcus aureus carriage identifies a subset of newly diagnosed granulomatosis with polyangiitis patients with high relapse rate. Rheumatology 2017, 56, 965–972. [Google Scholar] [CrossRef]
  18. Glasner, C.; de Goffau, M.C.; van Timmeren, M.M.; Schulze, M.L.; Jansen, B.; Tavakol, M.; van Wamel, W.J.B.; Stegeman, C.A.; Kallenberg, C.G.M.; Arends, J.P.; et al. Genetic loci of Staphylococcus aureus associated with anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitides. Sci. Rep. 2017, 7, 12211. [Google Scholar] [CrossRef]
  19. Gómez-Puerta, J.A.; Gedmintas, L.; Costenbader, K.H. The association between silica exposure and development of ANCA-associated vasculitis: Systematic review and meta-analysis. Autoimmun. Rev. 2013, 12, 1129–1135. [Google Scholar] [CrossRef]
  20. Hogan, S.L.; Cooper, G.S.; Savitz, D.A.; Nylander-French, L.A.; Parks, C.G.; Chin, H.; Jennette, C.E.; Lionaki, S.; Jennette, J.C.; Falk, R.J. Association of silica exposure with anti-neutrophil cytoplasmic autoantibody small-vessel vasculitis: A population-based, case-control study. Clin. J. Am. Soc. Nephrol. 2007, 2, 290–299. [Google Scholar] [CrossRef] [Green Version]
  21. Gatenby, P.A.; Lucas, R.M.; Engelsen, O.; Ponsonby, A.L.; Clements, M. Antineutrophil cytoplasmic antibody-associated vasculitides: Could geographic patterns be explained by ambient ultraviolet radiation? Arthritis Care Res. 2009, 61, 1417–1424. [Google Scholar] [CrossRef] [PubMed]
  22. Hutton, H.L.; Holdsworth, S.R.; Kitching, A.R. ANCA-Associated Vasculitis: Pathogenesis, Models, and Preclinical Testing. Semin. Nephrol. 2017, 37, 418–435. [Google Scholar] [CrossRef] [PubMed]
  23. Kumar, D.; Batal, I.; Jim, B.; Mendez, B.; Anis, K. Unusual case of levamisole-induced dual-positive ANCA vasculitis and crescentic glomerulonephritis. BMJ Case Rep. 2018, 2018, bcr-2018-225913. [Google Scholar] [CrossRef]
  24. Al-Hussain, T.; Hussein, M.H.; Conca, W.; al Mana, H.; Akhtar, M. Pathophysiology of ANCA-associated Vasculitis. Adv. Anat. Pathol. 2017, 24, 4. [Google Scholar] [CrossRef] [PubMed]
  25. Wilde, B.; Thewissen, M.; Damoiseaux, J.; van Paassen, P.; Witzke, O.; Cohen Tervaert, J.W. T cells in ANCA-associated vasculitis: What can we learn from lesional versus circulating T cells? Arthritis Res. Ther. 2010, 12, 1. [Google Scholar] [CrossRef]
  26. Wilde, B.; Thewissen, M.; Damoiseaux, J.; Knippenberg, S.; Hilhorst, M.; van Paassen, P.; Witzke, O.; Cohen Tervaert, J.W. Regulatory B cells in ANCA-associated vasculitis. Ann. Rheum. Dis. 2013, 72, 1416–1419. [Google Scholar] [CrossRef]
  27. Witko-Sarsat, V.; Daniel, S.; NoËl, L.H.; Mouthon, L. Neutrophils and B lymphocytes in ANCA-associated vasculitis. APMIS 2009, 117, 27–31. [Google Scholar] [CrossRef]
  28. Ballanti, E.; Chimenti, M.S.; Perricone, R. Small-Medium Vessel Vasculitides: Is the Complement System a Potential Forgotten Target? Isr. Med. Assoc. J. IMAJ 2015, 17, 2. [Google Scholar]
  29. Chen, S.F.; Wang, F.M.; Li, Z.Y.; Yu, F.; Zhao, M.H.; Chen, M. Plasma complement factor H is associated with disease activity of patients with ANCA-associated vasculitis. Arthritis Res. Ther. 2015, 17, 129. [Google Scholar] [CrossRef]
  30. Bantis, K.; Stangou, M.; Kalpakidis, S.; Nikolaidou, C.; Lioulios, G.; Mitsoglou, Z.; Iatridi, F.; Fylaktou, A.; Papagianni, A. Systemic complement activation in anti-neutrophil cytoplasmic antibody-associated vasculitis and necrotizing glomerulonephritis. Nephrology 2021, 26, 30–37. [Google Scholar] [CrossRef]
  31. Frangou, E.; Vassilopoulos, D.; Boletis, J.; Boumpas, D.T. An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): Implications for the pathogenesis and treatment. Autoimmun. Rev. 2019, 18, 751–760. [Google Scholar] [CrossRef]
  32. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  33. Nakazawa, D.; Masuda, S.; Tomaru, U.; Ishizu, A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat. Rev. Rheumatol. 2019, 15, 2. [Google Scholar] [CrossRef]
  34. Lee, K.H.; Kronbichler, A.; Park, D.D.Y.; Park, Y.; Moon, H.; Kim, H.; Choi, J.H.; Choi, Y.; Shim, S.; Lyu, I.S.; et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: A comprehensive review. Autoimmun. Rev. 2017, 16, 1160–1173. [Google Scholar] [CrossRef]
  35. Stangou, M.; Papagianni, A.; Bantis, C.; Liakou, H.; Pliakos, K.; Giamalis, P.; Gionanlis, L.; Pantzaki, A.; Efstratiadis, G.; Memmos, D. Detection of multiple cytokines in the urine of patients with focal necrotising glomerulonephritis may predict short and long term outcome of renal function. Cytokine 2012, 57, 120–126. [Google Scholar] [CrossRef]
  36. Berti, A.; Warner, R.; Johnson, K.; Cornec, D.; Schroeder, D.; Kabat, B.; Langford, C.A.; Hoffman, G.S.; Fervenza, F.C.; Kallenberg, C.G.M.; et al. Brief Report: Circulating Cytokine Profiles and Antineutrophil Cytoplasmic Antibody Specificity in Patients with Antineutrophil Cytoplasmic Antibody–Associated Vasculitis. Arthritis Rheumatol. 2018, 70, 1114–1121. [Google Scholar] [CrossRef] [PubMed]
  37. Khan, I.; Watts, R.A. Classification of ANCA-associated vasculitis. Curr. Rheumatol. Rep. 2013, 15, 383. [Google Scholar] [CrossRef] [PubMed]
  38. Luqmani, R.A.; Suppiah, R.; Grayson, P.C.; Merkel, P.A.; Watts, R. Nomenclature and classification of vasculitis—Update on the ACR/EULAR Diagnosis and Classification of Vasculitis Study (DCVAS). Clin. Exp. Immunol. 2011, 164 (Suppl. S1), 11–13. [Google Scholar] [CrossRef]
  39. Mukhtyar, C.; Lee, R.; Brown, D.; Carruthers, D.; Dasgupta, B.; Dubey, S.; Flossmann, O.; Hall, C.; Hollywood, J.; Jayne, D.; et al. Modification and validation of the Birmingham vasculitis activity score (version 3). Ann. Rheum. Dis. 2009, 68, 1827–1832. [Google Scholar] [CrossRef] [PubMed]
  40. Haris, Á.; Polner, K.; Arányi, J.; Braunitzer, H.; Kaszás, I.; Rosivall, L.; Kökény, G.; Mucsi, I. Simple, readily available clinical indices predict early and late mortality among patients with ANCA-associated vasculitis. BMC Nephrol. 2017, 18, 76. [Google Scholar] [CrossRef]
  41. Falk, R.J.; Jennette, J.C. ANCA disease: Where is this field heading? J. Am. Soc. Nephrol. 2010, 21, 745–752. [Google Scholar] [CrossRef] [Green Version]
  42. Xiao, H.; Heeringa, P.; Hu, P.; Liu, Z.; Zhao, M.; Aratani, Y.; Maeda, N.; Falk, R.J.; Jennette, J.C. Antineutrophil cytoplasmic autoantibodies specific for myeloperoxidase cause glomerulonephritis and vasculitis in mice. J. Clin. Investig. 2002, 110, 955–963. [Google Scholar] [CrossRef]
  43. Tervaert, J.W.C.; Damoiseaux, J. Antineutrophil cytoplasmic autoantibodies: How are they detected and what is their use for diagnosis, classification and follow-up? Clin. Rev. Allergy Immunol. 2012, 43, 211–219. [Google Scholar] [CrossRef] [PubMed]
  44. Kain, R.; Tadema, H.; McKinney, E.F.; Benharkou, A.; Brandes, R.; Peschel, A.; Hubert, V.; Feenstra, T.; Sengölge, G.; Stegeman, C.; et al. High prevalence of autoantibodies to hLAMP-2 in anti-neutrophil cytoplasmic antibody-associated vasculitis. J. Am. Soc. Nephrol. 2012, 23, 556–566. [Google Scholar] [CrossRef] [PubMed]
  45. Peschel, A.; Basu, N.; Benharkou, A.; Brandes, R.; Brown, M.; Rees, A.J.; Kain, R. Autoantibodies to hLAMP-2 in ANCA-negative pauci-immune focal necrotizing GN. J. Am. Soc. Nephrol. 2014, 25, 455–463. [Google Scholar] [CrossRef] [PubMed]
  46. Salama, A.D.; Pusey, C.D. Shining a LAMP on pauci-immune focal segmental glomerulonephritis. Kidney Int. 2009, 76, 15–17. [Google Scholar] [CrossRef]
  47. Thompson, G.E.; Fussner, L.A.; Hummel, A.M.; Schroeder, D.R.; Silva, F.; Snyder, M.R.; Langford, C.A.; Merkel, P.A.; Monach, P.A.; Seo, P.; et al. Clinical Utility of Serial Measurements of Antineutrophil Cytoplasmic Antibodies Targeting Proteinase 3 in ANCA-Associated Vasculitis. Front. Immunol. 2020, 11, 2053. [Google Scholar] [CrossRef]
  48. Fussner, L.A.; Hummel, A.M.; Schroeder, D.R.; Silva, F.; Cartin-Ceba, R.; Snyder, M.R.; Hoffman, G.S.; Kallenberg, C.G.; Langford, C.A.; Merkel, P.A.; et al. Factors Determining the Clinical Utility of Serial Measurements of Antineutrophil Cytoplasmic Antibodies Targeting Proteinase 3. Arthritis Rheumatol. 2016, 68, 1700–1710. [Google Scholar] [CrossRef] [PubMed]
  49. Yen, C.L.; Tian, Y.C.; Wu, H.H.; Tu, K.H.; Liu, S.H.; Lee, C.C.; Fang, J.T.; Yang, C.W.; Li, Y.J. High anti-neutrophil cytoplasmic antibody titers are associated with the requirement of permanent dialysis in patients with myeloperoxidase-ANCA-associated vasculitis. J. Formos. Med. Assoc. 2019, 118, 1408–1415. [Google Scholar] [CrossRef] [PubMed]
  50. Sethi, S.; Haas, M.; Markowitz, G.S.; D’Agati, V.D.; Rennke, H.G.; Jennette, J.C.; Bajema, I.M.; Alpers, C.E.; Chang, A.; Cornell, L.D.; et al. Mayo clinic/renal pathology society consensus report on pathologic classification, diagnosis, and reporting of GN. J. Am. Soc. Nephrol. 2016, 27, 1278–1287. [Google Scholar] [CrossRef]
  51. Berden, A.E.; Ferrario, F.; Hagen, E.C.; Jayne, D.R.; Jennette, J.C.; Joh, K.; Neumann, I.; Noël, L.H.; Pusey, C.D.; Waldherr, R.; et al. Histopathologic classification of ANCA-associated glomerulonephritis. J. Am. Soc. Nephrol. 2010, 21, 1628–1636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Van Daalen, E.E.; Wester Trejo, M.A.C.; Göçeroglu, A.; Ferrario, F.; Joh, K.; Noel, L.; Olgawa, Y.; Wilhemus, S.; Ball, M.J.; Honsova, E.; et al. Developments in the histopathological classification of ANCA-associated glomerulonephritis. Clin. J. Am. Soc. Nephrol. 2020, 15, 1103–1111. [Google Scholar] [CrossRef] [PubMed]
  53. Bjørneklett, R.; Sriskandarajah, S.; Bostad, L. Prognostic value of histologic classification of ANCA-associated glomerulonephritis. Clin. J. Am. Soc. Nephrol. 2016, 11, 2159–2167. [Google Scholar] [CrossRef] [PubMed]
  54. Hakroush, S.; Kluge, I.A.; Ströbel, P.; Korsten, P.; Tampe, D.; Tampe, B. Systematic histological scoring reveals more prominent interstitial inflammation in myeloperoxidase-anca compared to proteinase 3-anca glomerulonephritis. J. Clin. Med. 2021, 10, 1231. [Google Scholar] [CrossRef] [PubMed]
  55. Hauer, H.A.; Bajema, I.M.; van Houwelingen, H.C.; Ferrario, F.; Noel, L.; Waldherr, R.; Jayn, D.; Rasmussen, N.; Bruijn, J.; Hagen, E.; et al. Renal Histology in ANCA-Associated Vasculitis: Differences between Diagnostic and Serologic Subgroups. Kidney Int. 2002, 61, 80–89. [Google Scholar] [CrossRef]
  56. Eisenberger, U.; Fakhouri, F.; Vanhille, P.; Beafulis, H.; Mahr, A.; Guilevin, L.; Lesarve, P.; Noel, L. ANCA-negative pauci-immune renal vasculitis: Histology and outcome. Nephrol. Dial. Transplant. 2005, 20, 1392–1399. [Google Scholar] [CrossRef]
  57. Bantis, K.; Stangou, M.J.; Kalpakidis, S.; Nikolaidou, C.; Lioulios, G.; Mitsoglou, Z.; Iatridi, F.; Fylaktou, A.; Papagianni, A. Different Types of ANCA Determine Different Clinical Phenotypes and Outcome in ANCA-Associated Vasculitis (AAV). Front. Med. 2022, 8, 783757. [Google Scholar] [CrossRef]
  58. Paulsen, J.I.; Rudert, H. Manifestations of Primary Vasculitis in the ENT Region. Z. Fur Rheumatol. 2001, 60, 219–225. [Google Scholar] [CrossRef]
  59. Padoan, R.; Campaniello, D.; Felicetti, M.; Cazzador, D.; Schiavon, F. Ear, nose, and throat in ANCA-associated vasculitis: A comprehensive review. Vessel. Plus 2021, 5, 41. [Google Scholar] [CrossRef]
  60. Rahmattulla, C.; de Lind Van Wijngaarden, R.A.F.; Berden, A.E.; Hauer, H.A.; Floßmann, O.; Jayne, D.R.; Gaskin, G.; Rasmussen, N.; Noël, L.H.; Ferrario, F.; et al. Renal function and ear, nose, throat involvement in anti-neutrophil cytoplasmic antibody-associated vasculitis: Prospective data from the European Vasculitis Society clinical trials. Rheumatology 2014, 54, 899–907. [Google Scholar] [CrossRef]
  61. Solans-Laqué, R.; Fraile, G.; Rodriguez-Carballeira, M.; Caminal, L.; Castillo, M.J.; Martínez-Valle, F.; Sáez, L.; Rios, J.J.; Solanich, X.; Oristrell, J.; et al. Clinical characteristics and outcome of Spanish patients with ANCA-associated vasculitides Impact of the vasculitis type, ANCA specificity, and treatment on mortality and morbidity. Medicine 2017, 96, e6083. [Google Scholar] [CrossRef]
  62. Mohammad, A.J.; Mortensen, K.H.; Babar, J.; Smith, R.; Jones, R.; Nakagomi, D.; Sivasothy, P.; Jayne, D. Pulmonary involvement in antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis: The influence of ANCA subtype. J. Rheumatol. 2017, 44, 1458–1467. [Google Scholar] [CrossRef] [PubMed]
  63. Sharma, A.; Lakshman, A.; Nampoothiri, R.V.; Verma, R.; Rathi, M.; Naidu, G.S.; Pinto, B.; Sharma, K.; Dhir, V.; Nada, R.; et al. Pulmonary and Ear, Nose and Throat (ENT) Involvement in ANCA-Associated Vasculitis at Diagnosis-Experience from a Tertiary Care Centre in North India. J. Assoc. Physicians India 2017, 65, 40–47. [Google Scholar]
  64. Ungprasert, P.; Crowson, C.S.; Cartin-Ceba, R.; Garrity, J.A.; Smith, W.M.; Specks, U.; Matteson, E.L.; Makol, A. Clinical characteristics of inflammatory ocular disease in anti-neutrophil cytoplasmic antibody associated vasculitis: A retrospective cohort study. Rheumatology 2017, 56, 1763–1770. [Google Scholar] [CrossRef]
  65. Pagnoux, C. Updates in ANCA-associated vasculitis. Eur. J. Rheumatol. 2016, 3, 122–133. [Google Scholar] [CrossRef]
  66. Cohen Tervaert, J.W.; Stegeman, C.A.; Kallenberg, C.G.M. Novel Therapies for Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis. Curr. Opin. Nephrol. Hypertens. 2001, 10, 211–217. [Google Scholar] [CrossRef]
  67. Jayne, D. Treating vasculitis with conventional immunosuppressive agents. Clevel. Clin. J. Med. 2012, 79 (Suppl. S3), 46–49. [Google Scholar] [CrossRef]
  68. Geetha, D.; Shah, S. Place in therapy of rituximab in the treatment of granulomatosis with polyangiitis and microscopic polyangiitis. ImmunoTargets Ther. 2015, 4, 173–183. [Google Scholar] [CrossRef] [PubMed]
  69. Langford, C.A. Complications of Cyclophosphamide Therapy. Eur. Arch. Oto-Rhino-Laryngol. 1997, 254, 65–72. [Google Scholar] [CrossRef] [PubMed]
  70. De Groot, K.; Harper, L.; Jayne, D.R.; Flores Suarez, L.F.; Gregorini, G.; Gross, W.L.; Luqmani, R.; Pusey, C.D.; Rasmussen, N.; Sinico, R.A.; 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, 670–680. [Google Scholar] [CrossRef]
  71. Harper, L.; Morgan, M.D.; Walsh, M.; Hoglund, P.; Westman, K.; Flossmann, O.; Tesar, V.; Vanhille, P.; de Groot, K.; Luqmani, R.; et al. Pulse versus daily oral cyclophosphamide for induction of remission in ANCA-associated vasculitis: Long-term follow-up. Ann. Rheum. Dis. 2012, 71, 955–960. [Google Scholar] [CrossRef]
  72. Geetha, D.; Specks, U.; Stone, J.H.; Merkel, P.A.; Seo, P.; Spiera, R.; Langford, C.A.; Hoffman, G.S.; Kallenberg, C.G.; St Clair, E.W.; et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis with renal involvement. J. Am. Soc. Nephrol. 2015, 26, 976–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Unizony, S.; Villarreal, M.; Miloslavsky, E.M.; Lu, N.; Merkel, P.A.; Spiera, R.; Seo, P.; Langford, C.A.; Hoffman, G.S.; Kallenberg, C.M.; et al. Clinical outcomes of treatment of anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis based on ANCA type. Ann. Rheum. Dis. 2016, 75, 1166–1169. [Google Scholar] [CrossRef] [PubMed]
  74. Jones, R.B.; Cohen Tervaert, J.W.; Hauser, T.; Luqmani, R.; Morgan, M.D.; Peh, C.A.; Savage, C.O.; Segelmark, M.; Tesar, V.; van Paassen, P.; et al. Rituximab versus Cyclophosphamide in ANCA-Associated Renal Vasculitis. N. Engl. J. Med. 2010, 363, 211–220. [Google Scholar] [CrossRef] [PubMed]
  75. Jones, R.B.; Furuta, S.; Tervaert, J.W.C.; Hauser, T.; Luqmani, R.; Morgan, M.D.; Peh, C.A.; Savage, C.O.; Segelmark, M.; Tesar, V.; et al. Rituximab versus cyclophosphamide in ANCA-associated renal vasculitis: 2-year results of a randomised trial. Ann. Rheum. Dis. 2015, 74, 1178–1182. [Google Scholar] [CrossRef] [PubMed]
  76. Walsh, M.; Faurschou, M.; Berden, A.; Flossmann, O.; Bajema, I.; Hoglund, P.; Smith, R.; Szpirt, W.; Westman, K.; Pusey, C.D.; et al. Long-term follow-up of cyclophosphamide compared with azathioprine for initial maintenance therapy in anca-associated vasculitis. Clin. J. Am. Soc. Nephrol. 2014, 9, 1571–1576. [Google Scholar] [CrossRef]
  77. Karras, A.; Pagnoux, C.; Haubitz, M.; De Groot, K.; Puechal, X.; Tervaert, J.W.; Segelmark, M.; Guillevin, L.; Jayne, D. Randomised controlled trial of prolonged treatment in the remission phase of ANCA-associated vasculitis. Ann. Rheum. Dis. 2017, 76, 1662–1668. [Google Scholar] [CrossRef]
  78. De Joode, A.A.E.; Sanders, J.S.F.; Puéchal, X.; Groot, K.; Puechal, X.; Tervaert, J.W.C.; Segelmark, M.; Guillevin, L.; Jayne, D.; European Vasculitis Society. Long term azathioprine maintenance therapy in ANCA-associated vasculitis: Combined results of long-term follow-up data. Rheumatology 2017, 56, 1894–1901. [Google Scholar] [CrossRef]
  79. Guillevin, L.; Pagnoux, C.; Karras, A.; Khouatra, C.; Aumaître, O.; Cohen, P.; Maurier, F.; Decaux, O.; Ninet, J.; Gobert, P.; et al. Rituximab versus Azathioprine for Maintenance in ANCA-Associated Vasculitis. N. Engl. J. Med. 2014, 371, 1771–1780. [Google Scholar] [CrossRef]
  80. Charles, P.; Terrier, B.; Perrodeau, É.; Cohen, P.; Faguer, S.; Huart, A.; Hamidou, M.; Agard, C.; Bonnotte, B.; Samson, M.; et al. Comparison of individually tailored versus fixed-schedule rituximab regimen to maintain ANCA-associated vasculitis remission: Results of a multicentre, randomised controlled, phase III trial (MAINRITSAN2). Ann. Rheum. Dis. 2018, 77, 1144–1150. [Google Scholar] [CrossRef]
  81. Cartin-Ceba, R.; Golbin, J.M.; Keogh, K.A.; Peikert, T.; Sánchez-Menéndez, M.; Ytterberg, S.R.; Fervenza, F.C.; Specks, U. Rituximab for remission induction and maintenance in refractory granulomatosis with polyangiitis (Wegener’s): Ten-year experience at a single center. Arthritis Rheum. 2012, 64, 3770–3778. [Google Scholar] [CrossRef] [PubMed]
  82. McClure, M.; Jones, R.B. Treatment of relapses in ANCA-associated vasculitis. Clin. J. Am. Soc. Nephrol. 2019, 14, 967–969. [Google Scholar] [CrossRef] [PubMed]
  83. Tesar, V.; Hruskova, Z. Treatment of granulomatosis with polyangiitis and microscopic polyangiitis: Should type of ANCA guide the treatment? Clin. J. Am. Soc. Nephrol. 2020, 15, 1519–1521. [Google Scholar] [CrossRef] [PubMed]
  84. Mahr, A.; Katsahian, S.; Varet, H.; Guillevin, L.; Hagen, E.C.; Höglund, P.; Merkel, P.A.; Pagnoux, C.; Rasmussen, N.; Westman, K.; et al. French Vasculitis Study Group (FVSG) and the European Vasculitis Society (EUVAS). Revisiting the classification of clinical phenotypes of anti-neutrophil cytoplasmic antibody-associated vasculitis: A cluster analysis. Ann. Rheum. Dis. 2013, 72, 1003–1010. [Google Scholar] [CrossRef]
  85. Wójcik, K.; Biedroń, G.; Wawrzycka-Adamczyk, K.; Bazan-Socha, S.; Ćmiel, A.; Zdrojewski, Z.; Masiak, A.; Czuszyńska, Z.; Majdan, M.; Jeleniewicz, R.; et al. Subphenotypes of ANCA-associated vasculitis identified by latent class analysis. Clin. Exp. Rheumatol. 2021, 39 (Suppl. S129), 62–68. [Google Scholar] [CrossRef]
Figure 1. Geographic distribution of PR3-AAV, MPO-AAV and ANCA(−) AAV. Data obtained from Pearce et al. (‘Global, ethnic and geographic differences in the clinical presentations of anti-neutrophil cytoplasm antibody-associated vasculitis’). The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license (https://smart.servier.com (accessed on 1 August 2022)).
Figure 1. Geographic distribution of PR3-AAV, MPO-AAV and ANCA(−) AAV. Data obtained from Pearce et al. (‘Global, ethnic and geographic differences in the clinical presentations of anti-neutrophil cytoplasm antibody-associated vasculitis’). The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license (https://smart.servier.com (accessed on 1 August 2022)).
Life 12 01467 g001
Figure 2. Pathogenetic events in ANCA-associated vasculitis. A genetically determined background, combined with exposure to environmental factors, can disrupt homeostasis and lead to dysfunction of the immune system. Without the suppressive supervision of Tregs, there is an abnormal expansion of Th17 and effector CD25+ T-cell populations. Furthermore, Breg deficiency leads to insufficient suppression of proinflammatory T-cell populations. Dysregulated expression of the CD19 coreceptor by a subset of memory B cells has also been reported. The impaired function of immunity results in elevated amounts of C5a and circulating cytokines, with the capacity to prime neutrophils. Primed neutrophils expose on their surface PR3 and MPO antigens. ANCAs bind to these antigens and activate neutrophils. Activated neutrophils release NETs and positive regulators of the alternative complement pathway (properdin and factor B), promoting complement hyperactivation and C5a formation. The released NETs, ROS and proteases damage the endothelium and provoke vascular lesions.
Figure 2. Pathogenetic events in ANCA-associated vasculitis. A genetically determined background, combined with exposure to environmental factors, can disrupt homeostasis and lead to dysfunction of the immune system. Without the suppressive supervision of Tregs, there is an abnormal expansion of Th17 and effector CD25+ T-cell populations. Furthermore, Breg deficiency leads to insufficient suppression of proinflammatory T-cell populations. Dysregulated expression of the CD19 coreceptor by a subset of memory B cells has also been reported. The impaired function of immunity results in elevated amounts of C5a and circulating cytokines, with the capacity to prime neutrophils. Primed neutrophils expose on their surface PR3 and MPO antigens. ANCAs bind to these antigens and activate neutrophils. Activated neutrophils release NETs and positive regulators of the alternative complement pathway (properdin and factor B), promoting complement hyperactivation and C5a formation. The released NETs, ROS and proteases damage the endothelium and provoke vascular lesions.
Life 12 01467 g002
Figure 3. Classification of small-vessel vasculitis (SVV), based on immune deposits (CHCC 2012). SVV can be divided into ANCA-associated vasculitis, with few or no immune deposits, and immune complex SVV, with immunoglobin or complement deposits in vessel walls.
Figure 3. Classification of small-vessel vasculitis (SVV), based on immune deposits (CHCC 2012). SVV can be divided into ANCA-associated vasculitis, with few or no immune deposits, and immune complex SVV, with immunoglobin or complement deposits in vessel walls.
Life 12 01467 g003
Figure 4. Characteristics of ENT, lung and kidney involvement in PR3-AAV and MPO-AAV. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license (https://smart.servier.com (accessed on 1 August 2022)).
Figure 4. Characteristics of ENT, lung and kidney involvement in PR3-AAV and MPO-AAV. The Figure was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license (https://smart.servier.com (accessed on 1 August 2022)).
Life 12 01467 g004
Table 1. Definitions of the syndromic presentations of AAV, as established by the 2012 Revised International Chapel Hill Consensus Conference.
Table 1. Definitions of the syndromic presentations of AAV, as established by the 2012 Revised International Chapel Hill Consensus Conference.
CHCC 2012 NameCHCC 2012 Definition
Granulomatosis with polyangiitis (GPA)Necrotizing granulomatous inflammation, usually involving the upper and lower respiratory tract, and necrotizing vasculitis, predominantly affecting small-to-medium vessels. Necrotizing glomerulonephritis is common.
Microscopic polyangiitis (MPA) Necrotizing vasculitis, predominantly affecting small vessels. Necrotizing glomerulonephritis is very common. Pulmonary capillaritis often occurs. Granulomatous inflammation is absent.
Eosinophilic granulomatosis with polyangiitis (EGPA)Eosinophil-rich and necrotizing granulomatous inflammation, often involving the respiratory tract, and necrotizing vasculitis, predominantly affecting small-to-medium vessels. Associated with asthma and eosinophilia.
Single-organ AAVFor example, renal-limited vasculitis (RLV). Vasculitis in arteries or veins of a single organ, without any features indicating that it is a limited expression of a systemic vasculitis.
Table 2. Differences between PR3-AAV and MPO-AAV in terms of epidemiology, pathogenesis, histology and clinical manifestations.
Table 2. Differences between PR3-AAV and MPO-AAV in terms of epidemiology, pathogenesis, histology and clinical manifestations.
FeaturePR3-AAVMPO-AAV
Male-to-female ratioHigherLower
Age of onsetYoungerOlder
Geographic distributionPredominant in Northern EuropePredominant in Japan and China
GeneticsSNPs at the HLA-DP locus
HLA-DPB1*04 allele
SERPINA 1
PRTN3
SNPs at the HLA-DQ locus
Environmental factorsSt. Aureus
Low exposure to UV radiation
Vitamin D deficiency
Silica exposure
CytokinesIL-6, GM-CSF, IL-15, IL-18, CXCL8/IL-8, CCL-17/TARC, IL-18BP, sIL-2Ra, NGFb, BAFFsIL6R, sTNFRII, NGAL, sICAM-1
Ocular involvementMore commonLess common
ENT involvementVery commonLess common
Pulmonary involvementCentral airway disease
Nodular lesions
Interstitial fibrosis
Bronchiectasis
Alveolar hemorrhage
Renal histologyHigher fraction of healthy glomeruli
Frequently focal and crescentic class
Smaller fraction of healthy glomeruli
Frequently sclerotic class
Interstitial fibrosis
Tubular atrophy
RelapsesHigher riskLower risk
Renal survivalBetterWorse
Abbreviations: PR3, proteinase 3, MPO, myeloperoxidase, SNPs, single-nucleotide polymorphisms.
Table 3. Important clinical trials in the treatment of ANCA-associated vasculitis.
Table 3. Important clinical trials in the treatment of ANCA-associated vasculitis.
Clinical TrialObjectiveResults
CYCLOPSPulse CTX versus daily oral CTX for remission inductionPulse CTX maintains the efficacy of oral CTX at a smaller cumulative dose
CYCLOPS long-term follow upLong-term outcomes of the CYCLOPS studyMPO-ANCA patients respond equally to both regimes
PR3-ANCA patients are at a higher risk of relapse when treated with CTX in pulses
RAVERTX versus oral CTX for remission inductionRTX matches the efficacy of oral CTX
RAVE post hoc analysisEffect of ANCA specificity on treatment responsePR3-ANCA patients respond better to RTX than to oral CTX
Comparable efficacy in MPO-ANCA patients
RITUXVASRTX versus CTX in pulses for remission inductionRTX matches the efficacy of CTX in pulses
Two-year follow up of the RITUXVAS studyLong-term outcomes of the RITUXVAS studyOutcome of death, ESRD and relapse did not differ significantly between the two regimen groups
CYCAZAREMOptimal time interval to switch from CTX to AZT for remission maintenanceEarly CTX replacement leads to more relapses, but the difference is not statistically important. PR3-ANCA patients relapse more often than MPO-ANCA patients after CTX suspension
REMAINEfficacy of two different durations of an AZT-based maintenance therapyProlonged maintenance therapy, beyond 24 months, reduces relapse risk and improves survival
IMPROVEMMF versus AZT for remission maintenanceMMF is less effective than AZT for remission maintenance
MAINRITSANRTX versus AZT for remission maintenanceRTX is superior to AZT for maintenance of remission
MAINRITSAN2Tailored, based on trimestral biological parameters versus fixed-schedule RTX infusionsRelapse rates were similar between the two administration regimens
MAINRITSAN2 post hoc analysisEffect of omitting RTX administration at day 14Relapse-free survival rates did not differ significantly
Rituximab for remission induction and maintenance in refractory GPAEfficacy of RTX for maintenance of long-term remission in refractory GPARTX is effective for long-term maintenance of remission in PR3-ANCA patients. Monitoring B-cell levels can help to predict and prevent a relapse
Abbreviations: CTX, cyclophosphamide, AZT, azathioprine, RTX, rituximab, MMF, mycophenolate mofetil.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Konstantouli, A.M.; Lioulios, G.; Stai, S.; Moysidou, E.; Fylaktou, A.; Papagianni, A.; Stangou, M. Type of ANCA May Be Indispensable in Distinguishing Subphenotypes of Different Clinical Entities in ANCA-Associated Vasculitis. Life 2022, 12, 1467. https://doi.org/10.3390/life12101467

AMA Style

Konstantouli AM, Lioulios G, Stai S, Moysidou E, Fylaktou A, Papagianni A, Stangou M. Type of ANCA May Be Indispensable in Distinguishing Subphenotypes of Different Clinical Entities in ANCA-Associated Vasculitis. Life. 2022; 12(10):1467. https://doi.org/10.3390/life12101467

Chicago/Turabian Style

Konstantouli, Afroditi Maria, Georgios Lioulios, Stamatia Stai, Eleni Moysidou, Asimina Fylaktou, Aikaterini Papagianni, and Maria Stangou. 2022. "Type of ANCA May Be Indispensable in Distinguishing Subphenotypes of Different Clinical Entities in ANCA-Associated Vasculitis" Life 12, no. 10: 1467. https://doi.org/10.3390/life12101467

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop