Download PDF
Review  |  Open Access  |  12 Jul 2021

Pathogenesis and biomarkers in ANCA-associated vasculitis

Views: 3481 |  Downloads: 969 |  Cited:   0
Vessel Plus 2021;5:44.
10.20517/2574-1209.2021.66 |  © The Author(s) 2021.
Author Information
Article Notes
Cite This Article

Abstract

Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs) are a group of systemic autoimmune diseases associated with serum ANCA positivity that affect small to medium vessels with inflammation and endothelial injury. This group includes several diseases: granulomatosis with polyangiitis (GPA), microscopic polyangiitis, eosinophilic GPA, and drug-induced AAV. A few AAVs are ANCA negative, but this form has decreased with the increase in detection methods. Different genetic, epigenetic, and environmental risk factors contribute to the pathogenesis of AAV. ANCA’s role in the origin of vasculitis has led to a better grasp of the disease. Research has also improved the treatment, more precisely to tune its intensity, translating into better outcomes. However, there is still a gap to be filled with new potential and testable biomarkers for diagnosis, disease activity, and prognosis, which we discuss here.

Keywords

Pathogenesis, ANCA, ANCA-associated vasculitis, biomarkers

INTRODUCTION

Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAVs) are a group of systemic autoimmune diseases that affect small to medium vessels with inflammatory and endothelial injury. In the early 1980s, the first case report mentioning ANCA presence in the serum of patients with segmental necrotizing glomerulonephritis triggered a new chapter in immune-mediated diseases[1]. The discovery of the major antigens targets of ANCAs [myeloperoxidase (MPO) and proteinase 3 (PR3)] led to the creation of an inclusive group of AAVs. Classification criteria based on clinical findings and tissue biopsy, if possible or reasonable, have not always been widely accepted and new scores are being proposed[2,3].

The AAV group incorporates at least four particular diseases: granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), eosinophilic GPA (EGPA; formerly Churg-Strauss syndrome), and drug-induced AAV[4].

GPA is a necrotizing small-vessel vasculitis characterized by the involvement of the respiratory tract with granulomatous inflammation. These patients are mainly PR3-ANCA positive. In contrast, MPA preferably targets renal glomeruli small vessels and presents a histological pattern of necrotizing vasculitis normally lacking granulomatous inflammation. The serum of these patients commonly presents MPO-ANCA positivity.

The EGPA histological pattern is similar to GPA, except its main feature of eosinophil-rich reaction that involves small- to medium-sized vessels. Patients with this disease typically have associated adult-onset asthma and allergic sinusitis, eosinophilia, tissue infiltrating eosinophils, and around 50% of the cases have MPO-ANCA positivity.

Drugs such as propylthiouracil, hydralazine, or cocaine induce ANCA production and generate drug-induced AAV. The clinical features of this particular spectrum of AAV have a wide and global variation according to ethnic differences[5]. More commonly, MPA- and MPO-ANCA positivity is observed in East Asian countries, while GPA- and PR3-ANCA is more noticeable in Western countries[6].

Worldwide identification of these diseases prompted international guidelines and treatment management for remission and maintenance[7-9]. However, methods for the monitoring of AAVs activity are still lacking. In this review, we characterize the current knowledge regarding AAV’s risk factors and pathogenesis and subsequently review newly discovered biomarkers for its diagnosis, monitoring, and prognosis.

RISK FACTORS OF AAV

A multiplicity of risk factors for AAV has been described in the literature, as summarized in Table 1.

Table 1

Anti-neutrophil cytoplasmic antibody-associated vasculitis risk factors

Risk factorEvidence
GeneticsSignificant association HLA-DP locus and GPA; DQ loci and MPO-ANCA; HLA-DP allele and GPA
SERPINA-1, α-1-antitrypsin allele deficiency, proteinase 3 gene (PR3) polymorphism and variant increasing PTPN22 activity
EpigeneticsMPO and PRTN3 promoter methylation
ExposomeSeasonalityDiscrepant results in literature
Onset observed in autumn and winter months
Latitude and UV radiationIncreased prevalence of MPA and GPA in hot and cold countries, respectively
Higher latitudes correlate with higher GPA incidence
InfectionsExposure to toxic shock syndrome toxin-1
Nasal S. aureus colonization related to relapse rates
PollutionSilica2.5-fold increased risk, even higher for patients with renal involvement, GPA or MPA
FarmingGPA associated with 12 months prior-visits and significant livestock exposure
Gardening, namely digging, mowing and planting
Other pollutantsExposure to carbon monoxide, hydrocarbon and high organic solvent
Drugse.g., propylthiouracil, methimazole, hydralazine, minocycline or cocaine
SmokingWeak correlation

Genetics

Even though familiar reports of AAV are rare[10], as for other autoimmune disorders, genome-wide association studies (GWAS) have been performed to further identify alleles related with patterns of susceptivity or resistance to AAV. Among these, the strongest associations were found for major histocompatibility complex class II (MHC II) and HLA gene locus on chromosome 6.

The differences in HLA are related to the ethnicity of the population and the appearance of certain circulating antibodies rather than the clinical appearance[11]. A GWAS conduced in the UK found strong associations between HLA-DPB1*04 and PR3-ANCA (OR = 7.03), MPO-ANCA and DQ loci (OR = 0.65), and HLA-DP allele and GPA (OR = 5.39)[11]. In Japan, population studies linked HLA-DRB1*09:01 with MPO-ANCA (OR = 1.57) and MPA (OR = 1.56). Curiously, HLA-DRB1*13:02 was suggested as a protective factor against AAV[12].

Besides MHC genes, several single-nucleotide polymorphisms (SNP) associated with the AAVs have been found. The variant that increases the activity of PTPN22, which negatively regulates IL-10 (an immunosuppressive cytokine), increases the likelihood of PR3-ANCA (OR = 1.63). Oppositely, SNPs related to SERPINA1, PRTN3, and SEMA6A showed profiles protective against AAV[11].

Epigenetics

Gene expression regulation of MPO and PRTN3 genes were linked to DNA methylation and histone 3 lysine 27 (H3K27me3). MPO and PRTN3 promoter methylation are negatively correlated with patients with active AAV and increase during remission of the disease[13]. H3K27me3 trimethylation reduction was also associated with active disease by expressing aberrant MPO and PRTN3 genes[14].

Exposome

The initial report of AAV was coupled with an infectious disease[1]. More recently, AAVs have been shown to be triggered by infectious agents, namely after exposure to toxic shock syndrome toxin-1, a toxin secreted by Staphylococcys aureus, and relapses have also been associated with higher nasal S. aureus colonization[15,16].

Many other factors appear to be related with higher risk for AAVs. Seasonality shows discrepant results in the literature, but AAVs, namely GPA, seem to have a more prevalent onset in autumn and winter months. The prevalence of AAV subtypes also seems linked with temperature, latitude, and UV radiation[17,18].

As mentioned above, some drugs can precipitate AAV; thus, drug indications need to be weighed when studying the correlation between drugs and the risk for AAVs. Alopurinol seems to be the only drug that conferred a greater risk for developing GPA[19].

The role of airborne particles has also been studied, with multiple approaches suggesting a higher disease risk when being exposed to certain pollutants, namely silica, carbon monoxide, hydrocarbons, and high organic solvents and particles associated with farming and gardening activities. As for smoking, there is a weak correlation to higher risk for developing the disease[17,20-22].

ANCA PATHOGENICITY

ANCA patterns

Two major types of ANCA can be detected by indirect immunofluorescence of ethanol-fixated neutrophils. Perinuclear ANCAs (p-ANCAs) stain around the nucleus and are mainly composed of myeloperoxidase. Conversely, some ANCAs stain diffusely in the cytoplasm, namely cytoplasmatic ANCAs (c-ANCAs), of which the most important is PR3. Other “minor” ANCAs have been described: against α-enolase, azurocidin, bactericidal permeability-increasing protein (BPI), cathepsin G, elastase, defensin, lactoferrin, lysosome-associated membrane glycoprotein 2 (LAMP2), and moesin. These antibodies are mainly p-ANCAs and are rarely associated with vasculitis. Nowadays, enzyme-linked immunosorbent assay (ELISA) tests detect MPO and PR3 instead of p-ANCAs and c-ANCAs, which correspond to the staining pattern[15].

ANCA production

Neutrophils participate in the innate immune defense. These cells form networks of extracellular fibers, primarily composed of DNA from neutrophils, which bind pathogens[23], the so-called neutrophil extracellular traps (NETs). An imbalance in NETs’ production is known to participate in ANCA production. These traps are degraded by serum DNAse I. Nevertheless, in some circumstances, as in the case of propylthiouracil administration, NETs become resistant to DNAse I. The overexposure to NETs promotes the production of autoantibodies against neutrophils. Meanwhile, the traps are formed, and the content of neutrophilic granules, including PR3 and MPO, becomes mixed with chromatin fibers, which bind to DNA. By this physiological step, the antigenicity of PR3 and MPO might suffer modifications[24]. However, only a second step that prevents a complete degradation of NETs seems to be specific to some autoimmune diseases, such as AAVs and systemic lupus erythematosus[24,25]. This impaired NET degradation coincides with the lower serum DNAse I activity observed in patients with AAV when compared to healthy individuals[26]. Moreover, a later step triggers the autoimmune reaction by presenting misfolded proteins/antigens, in which cryptic epitopes are then recognized in class II MHC molecules by the immune system[27].

PATHOGENESIS OF AAV

Facing a bacterial or viral infection, dendritic cells produce tumor necrosis factor (TNF)-β and interleukin-6 (IL-6). These inflammatory markers stimulate the differentiation of T cells into T helper 17 (Th17). In turn, Th17 cells produce IL-17, an important cytokine that can induce the production of TNF-α or IL-1β by macrophages. These proinflammatory cytokines then prime the innate immune system, namely neutrophils and the formation of ANCAs. Additionally, the autoreactive response is sustained by imbalances of T regulatory cells (Treg) that regulate the differentiation of Th17 and are associated with increased risk of AAV[28,29].

The priming of the neutrophil also occurs by the complement system, namely C5a by the altern complement pathway, as described in animal models[30], which is related to hypercoagulability states in patients with AAV due to tissue factor release[31].

Upon priming, neutrophils express surface ANCA antigens. These ANCAs, besides binding to the corresponding antigens, also bind to the Fcγ receptor in neutrophils. This positive feedback induces an excessive activation of these neutrophils, promoting cytokine production and reactive oxygen species and lytic enzymes release[32], further incrementing the inflammatory status. ANCA-stimulated neutrophils release NETs that contribute to the local inflammatory response and epithelial damage [Figure 1]. At the same time, more neutrophils are activated either directly by NETs or indirectly by circulating ANCA.

Pathogenesis and biomarkers in ANCA-associated vasculitis

Figure 1. The inflammation cascade of anti-neutrophil cytoplasmic antibody (ANCA)-antibody associated vasculitis. ANCAs produced by plasma cells bind to myeloperoxidase and proteinase 3 produced by activated neutrophils (mediated by factors such as TNF-α or IL-1β). The neutrophil activation stimulates more neutrophils, leading to the production of extracellular traps (NET) and reactive oxygen species, leading to tissue damage, granulomatosis, and necrosis (image created in biorender.com). MPO: Myeloperoxidase; PR3: proteinase 3; NETs: neutrophil extracellular traps; ROS: reactive oxygen species; TNF-α: tumor necrosis factor α; IL-1β: interleukin-1β.

B cells are also activated by CD4+ T presenting cells, through the presentation of antigens resulting from NETs[33]. Another important mechanism may be by the release of B-cell-activating factor (BAFF) or B-lymphocyte stimulator (BLyS) from activated neutrophils[33].

In GPA, chronic exposure to Staphylococcus aureus by nasal mucosa colonization might be the pathogenic trigger[34], by priming a local innate immune response that, under the right circumstances, may end up in AAV development. In the case of EGPA, which features eosinophilia, a vascular endothelial cell release of eotaxin 3 has been implicated in the tissue infiltration of eosinophils. These eosinophils secrete major basic proteins, eosinophilic cationic proteins, and neurotoxins that destroy the tissue[35]. Despite this particularity, the mechanism of MPO-ANCA production in these patients is still undisclosed.

BIOMARKERS IN AAV

Finding non-invasive markers capable of diagnosis and predicting disease activity, prognosis, and therapeutic options is a well desired goal for numerous studies related to AAV. As such, many studies have focused on the search for practical non-invasive markers to track AAV, as discussed below and summarized in Table 2.

Table 2

Anti-neutrophil cytoplasmic antibody-associated vasculitis biomarkers and their usefulness

ApproachBiomarkerFunctionDescription
DiagnosisPR3-ANCA and MPO-ANCADistinguish from healthy patients and between AAV subtypesPresent in patients with GPA and MPA subtypes, respectively
cfDNADistinguish between AAV subtypesIncreased levels of cfDNA in PR3-ANCA GPA patients compared to
sCD163Distinguish from healthy patientsHigher urinary levels of sCD163 in patients with AAV
Eotaxin-3, IgG4 and CCL17/TARCDistinguish between AAV subtypesIncreased levels of Eotaxin-3, IgG4 and CCL17/TARC in EGPA compared to other AAV subtypes
12-HETEDistinguish between AAV subtypesHigher exhaled breath concentrations of 12-HETE in EGPA patients than healthy patients
PrognosisRecognizable N-terminus of MPO heavy chainEvaluate disease prognosisRecognizable N-terminus of MPO heavy chain are more prone to develop severe disease
C3Evaluate disease prognosisLow serum C3 levels is associated with poorer outcomes
Disease activityPTX3+ and HMGB+Distinguish disease activity statusConcentrations of PTX3+ and HMGB+ were higher in active AAV compared to remission
cfDNADistinguish disease activity statusConcentration levels are higher in disease activity phases namely in PR3-ANCA positive GPA
MCP-1, AGP, KIM-1, NGALDetect renal flaresUrinary levels of MCP-1, AGP, KIM-1, NGAL are increased in renal flares of AAV
sCD163Distinguish disease activity statusHigher urinary levels of sCD163 in disease activity phases
ESR, CRP and CalprotectinDistinguish disease activity statusHigher levels of ESR, CRP and Calprotectin in disease activity phases
Eotaxin-3, IgG4 and CCL17/TARCDistinguish disease activity statusIncreased levels of Eotaxin-3, IgG4 and CCL17/TARC in disease activity phases
sRAGEDistinguish disease activity statusAssessing mild/limited disease activity
EMPs and CECsDistinguish disease activity statusEMPs and CECs levels positively correlate with disease activity
CD25+ T-cells
Th17 cells
Distinguish disease activity statusCD25+ T-cells inversely correlate with disease activity
Th17 cells positively correlate with disease activity
T follicular helper (Tfh) cells
Tfh2/Tf1 ratio
Distinguish disease activity statusTfh cells positively correlate with disease activity
Tfh2/Tfh1 ratio shift
Bb, C3a, C5a and soluble C5b-9Distinguish disease activity statusBb, C3a, C5a and soluble C5b-9 urinary levels positively relate with disease activity.
Relapse riskPR3-ANCAEvaluate relapse riskIncreased PR3-ANCA levels positively relate with higher relapse risk
CalprotectinEvaluate relapse riskElevated calprotectin levels in patients who relapsed
EPCsEvaluate relapse riskReduced levels of EPCs positively relate with higher relapse risk
B-cellsEvaluate relapse riskIncomplete B-cell depletion and B-cell repopulation after rituximab treatment relate with higher relapse rate
CD8+ T-cellsEvaluate relapse riskCD8+ T-cells levels positively relate with higher relapse risk
Response to treatmentPR3-ANCAEvaluate response to treatmentPR3-ANCA positive respond better to treatment with Rituximab

ANCA

The value of ANCA as a diagnostic marker is mainly irrefutable. Occasional cases of “ANCA-negative” AAV are acknowledged, decreasing by the day with ANCA testing methodology improvement[36,37]. False-negative results can still be found because in some cases ANCA binds to the circulating ceruloplasmin[38].

The value of ANCA goes beyond its simple diagnostic value. The ANCA-positive patient subtypes, namely PR3-ANCA and MPO-ANCA, differ from one another regarding genetic basis, epidemiology, clinical manifestations, histological findings, response to therapy, and pathogenesis. These facts sustain the reason to distinguish between PR3-ANCA and MPO-ANCA patients, meaning GPA (commonly associated with PR3-ANCA) and MPA (commonly associated with MPO-ANCA)[36,39].

Whereas the ANCA diagnostic value is no longer under doubt, its role as a disease activity marker has been the subject of frequent studies with varying outcomes[40]. Discrepant results regarding disease activity have been a matter of discussion in the literature[41-43]. This misalignment in the results can be interpreted by differences in the epitopes and affinities of ANCAs. Patients with a recognizable N-terminus of MPO heavy chain are more prone to develop a severe disease[44], while patients with low-affinity despite high titers of MPO-ANCA have a lower vasculitic disease activity[45]. Some clinical studies show a direct link between an increment in PR3-ANCA levels during complete remission and an increased risk of relapse[46,47], while others find a direct link between PR3-ANCA-positive patients and relapse probability compared to MPO-ANCA patients[48,49].

The role of ANCA subtypes in predicting response to treatment is also a subject under scrutiny. Some studies suggest that adult PR3-ANCA-positive patients respond better to rituximab than to conventional induction/remission maintenance treatment with cyclophosphamide and azathioprine, leading to the belief that ANCA serotype may guide the type of treatment in AAV[50].

Biomarkers derived from neutrophil activation - NMPs and NETs

Neutrophil microparticles (NMPs) are membrane vesicles that induce endothelial damage in AAV, whereas NETs, composed of DNA, histones, and neutrophil proteins and released by ANCA-stimulated neutrophils, contain antigenic components including PR3 and MPO[51].

Removal of NMPs by filtration abolished a pathological trigger connected to endothelial activation, suggesting a target for therapeutic plasma exchange in AAV. Despite the suggestion, clinical utility remains unvalidated. A recent study concluded that MPO patients’ NMPs express higher levels of pentraxin-3 (PTX3), high mobility group box 1 protein (HMGB1), and tumor necrosis factor-like weak inducer of apoptosis (TWEAK) when compared to healthy controls[52]. Concentrations of PTX3+ and HMGB1+ were significantly higher in active AAV patients compared to those in remission[52]. PTX3 serum levels were strongly correlated with the Birmingham vasculitis activity score[52].

Moreover, Lange et al.[53] detected a significant increase in serum circulating free DNA (cfDNA) levels in PR3-ANCA GPA patients compared to EGPA and concluded that there is an association between concentration of cfDNA and disease activity. These findings suggest that abnormal formation and/or insufficient clearance of NETs may contribute to increase the levels of cfDNA in GPA. The detection of cfDNA levels or NETs may serve as a marker of disease activity in AAV, namely PR3-ANCA-positive GPA[53,54].

Urinary biomarkers

Urinary biomarkers could be ideal for the non-invasive monitoring of disease activity and renal involvement.

Lieberthal et al.[55] found that urinary levels of monocyte chemotactic protein 1 (MCP-1), α-1-acid glycoprotein (AGP), kidney injury molecule 1 (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL) presented statistically significant increases during renal flares among patients with AAV. Of those, MCP-1 chemokine granted the best discrimination between active renal disease and remission[55]. MCP-1 is in fact the most promising urinary biomarker for AAV still under research, with multiple studies suggesting usefulness in the assessment of vasculitis and monitoring treatment[55-57].

Urinary soluble CD163 was presented as an even more promising biomarker of renal vasculitis than urinary MCP-1[58]. CD163 is a soluble form of a high affinity scavenger receptor for the hemoglobin-haptoglobin receptor complex and functions as an innate sensor for bacteria. Patients with small vessel vasculitis (which includes GPA, MPA, and EGPA) had markedly higher urinary sCD163 levels than patients in remission, disease controls, or healthy controls[58]. However, serum sCD163 levels failed to distinguish infections from active disease, which may limit its use[59].

Inflammatory markers

Traditional inflammatory markers such as ESR (erythrocyte sedimentation rate) or CRP (C-reactive protein) are non-specific markers for AAV.

Monach et al.[60] evaluated whether these markers were able to distinguish severe AAV [BVAS for Wegener’s granulomatosis (BVAS/WG) ≥ 3 at screening] from remission. All subjects with severe active vasculitis at screening were followed during remission at Month 6, of whom 24 out of 28 showed significant decline[60]. Although they are not specific, traditional inflammatory markers correlate with the activity of the disease.

Eosinophilia is the hallmark of EGPA; however, the number of circulating eosinophils may not be an adequate biomarker for active EGPA since the eosinophil count usually drops dramatically and rapidly after treatment with glucocorticoids. Indeed, the treatment effect, mainly glucocorticoids, may directly cause the mispresenting comparison between active and inactive EGPA. Eotaxin-3, immunoglobulin G4 (IgG4), and CC chemokine ligand 17 (CCL17/TARC) levels were higher in active EGPA compared to healthy controls, inactive EGPA, and/or other diseases featuring vasculitis or hypereosinophilia[35,61,62]. Interestingly, eotaxin-3 levels were lower in hypereosinophilic syndromes than EGPA, underpinning a potential diagnosis use[35,61].

Pepper et al.[63] suggested calprotectin as a potential disease biomarker in patients with AAV by showing that patients with AAV had higher monocyte and neutrophil cell surface calprotectin expression than healthy controls and that its levels increased following treatment withdrawal and were significantly elevated in patients who relapsed.

12-Hydroxyeicosatetraenoic acid (12-HETE) measurement in exhaled breath concentrates (EBC) is a very different approach to distinguishing EGPA from asthma or hypereosinophilic syndromes. Szczeklik et al.[64] identified markedly higher concentrations of 12-HETE in EBC of EGPA than of other compared diseases, even though 19 out of 23 patients were receiving glucocorticoids and one third were clinically in remission.

Clinical trials also provide promising markers, such as MMP-3, TIMP-1, and CXCL13 (BCA-1)[60], which need further investigation with proper endpoints. sRAGE (serum advanced glycation end products) is another particular biomarker that may reflect the burden of granulomatous inflammation in GPA[65] and therefore be useful for assessing mild/limited disease activity.

Endothelial damage biomarkers

Two important biomarkers of endothelial injury were studied in AAV: endothelial microparticles (EMPs) and circulating detached mature endothelial cells (CECs).

EMPs are complex vesicular structures released from activated or apoptotic endothelial cells that play a singular role in inflammation, coagulation, endothelial function, and angiogenesis. Their dysregulation disturbs the vascular homeostasis, contributing to the progression of vascular diseases, such as vasculitis[66,67]. For this reason, research on EMPs identified a positive correlation with disease activity when compared with patients in remission[68]. These same EMPs may generate excess thrombin and thrombotic complications, namely in children with systemic vasculitis[69] and cerebral vasculitis of the young[70].

CECs have also been described as a biomarker of disease activity in vasculitis[71]. These are necrotic, highly activated endothelial cells that detach from the vessel wall and positively correlate with vasculitis in adults and children[67,71]. Upon endothelial lesion, bone marrow-derived endothelial progenitor cells (EPCs) increase in order to perform endothelial repair. These events have been described to be predictive of early relapse in adults with AAV, in whom a reduced number of circulating EPCs has been observed[72].

Von Willebrand factor (VWF) is yet another biomarker of disease to be confirmed in adult AAV patients. This plasma protein, synthesized primarily by megakaryocytes and endothelial cells, mediates platelet aggregation and adhesion. VWF levels increase in response to endothelial injury or activation, and it has been described as a biomarker of disease activity in childhood CNS vasculitis that was not confirmed in adults[70,73].

B cell, T cell, and cytokines

B-cell subset populations have drawn study interest, with data from observational studies suggesting that incomplete B-cell depletion and B-cell repopulation after rituximab treatment is associated with a significantly higher relapse rate[74,75].

B-cell subset populations of interest include regulatory B cells (Bregs), such as CD5+ cells[40]. Measurement of Bregs showed initial promise with a lower CD5+ B-cell count correlating with active disease, but a subsequent analysis from the RAVE study found that this count was not predictive of disease relapse, severity, or treatment failure[76,77].

In some studies, elevated levels of B-cell activating factor (BAFF) have been found in patients with AAV active disease, with a corresponding fall after treatment. However, BAFF levels are affected by corticosteroid treatment, rendering it an inadequate biomarker in AAV[78].

T-cell involvement in AAV was previously discussed. CD25 and CD28 T-cell markers were already correlated with disease activity. CD25+ marker on T cells was inversely related to disease activity, however it is not clear if its population reflects a cause or effect of the disease[39]. CD28+ T cells are associated with a high risk of relapse, displaying markers associated with T-cell survival and memory T-cell levels; thus, the CD8+ T-cell profile might be able to identify a group of AAV patients more likely to relapse[39]. Another subset of T cells, Th17 cells, has been identified as a potential driver of the disease. Th17 cells seem to be autoantigen-specific and are also spotted in renal lesions of AAV patients[79]. von Borstel et al.[80] analyzed CD19+CD24hiCD38hi B cells from the peripheral blood of AAV patients in remission and healthy controls. They found a negative correlation between Th17 cells and Breg and concluded that Th17 cells in AAV patients are at least partially controlled by Breg, highlighting another population that may be responsible for the disease and a target for its control[80]. Indeed, targeting B cells by the use of rituximab has shown a sustained remission at Month 28 when compared with other maintenance options[81,82]. However, as discussed above, B-cell activity has not been predictive of disease relapse. Other research groups found that an imbalance of Th17 and regulatory T (Treg) cells may also be of potential use to monitor AVV patients with renal involvement[83]. An increase of Th17 cells and a decrease of Treg cells, along with a downward trend for IL-2 and IL-4 and an upward one for IL-6, IL-10, TNF-α, IFN-γ, and IL-17A, were observed in AVV patients[83].

T follicular helper (Tfh) cells and T follicular regulatory (Tfr) cells balance is critical for humoral immune responses and relevant in autoimmune diseases[84]. AVV patients present increased circulating Tfh cells (CD4+CXCR5+CD25CD127interm-hi), decreased Tfr cells (CD4+CXCR5+CD25+CD127lo-interm), and elevated Tfh/Tfr ratios compared with healthy control[84]. Moreover, this study observed a Tfh2/Tfh1 shift and increased plasma IL-21 level associated with AAV and disease activity[84]. This suggests Tfh phenotype as a promising biomarker for AVV disease activity.

Complement

Alternative complement pathway activation is a fundamental way of developing disease. Gou et al.[85] found that urinary levels of Bb, C3a, C5a, and soluble C5b-9 were significantly higher in active disease.

Recently, the role of C5a in ANCA-induced neutrophil activation was shown, relating the generation of C5a by supernatants from ANCA-stimulated neutrophils with the role of C5a mediated effects via its specific receptor, allowing neutrophils to generate ROS in response to ANCA[31]. It was demonstrated that the lack of C5a receptor induces resistance to ANCA-induced disease, namely experimental anti-MPO vasculitis with the significant attenuation of the neutrophil glomerular influx and lower albuminuria, showing a potential therapeutic target in AAV[86].

In the first AAV human study of C5a, Yuan et al.[87] demonstrated higher levels of C5a in patients with active AAV compared with AAV in remission. Similarly, plasma levels of fragment Bb were significantly higher in active AAV than in patients in remission or healthy controls. Those levels positively correlated with the number of crescents on renal histology, vasculitis activity score, and serum inflammatory markers[88]. More recently, the ADVOCATE study group investigated an C5 receptor inhibitor in patients with ANCA-associated vasculitis-avacopan, which proved noninferior but not superior to prednisone taper with respect to remission at Week 26 and was superior to prednisone taper with respect to sustained remission at Week 52[89]. This result represents a role for C5 as a possible biomarker, as described above in the pathogenesis of the disease.

Finally, Manenti et al.[90] reported that low serum C3 levels at diagnosis is associated with poorer patient and renal outcomes in AAV patients; however, there were no significant association between serological and pathohistological phenotypes and serum C3 levels.

CONCLUSION

In this review, we discuss an updated understanding of the pathogenesis and novel biomarkers of AAV. The knowledge gap in the pathophysiology and follow-up of AAV diseases has raised research interest. Innovations in methodology, such as modern “omics”, and new discoveries from other fields have incremented the AAV pathogenesis perception. Genetic, epigenetic, and environmental factors identification, along with the role of neutrophils, other immune cells, and humoral factor contributions, have now contributed to another milestone in AAV research. However, disease activity monitoring, treatment, and relapse prediction are not yet effective. Our most recent knowledge on the pathogenesis is identifying pivotal players in this disease and engendering new biomarkers soon to be studied in clinical settings. However, the demonstration of either the very high sensitivity or the very high specificity of new biomarkers has not yet reached clinically relevant outcomes.

Future collaborative research and interactions between basic and clinical research would be a good strategy for the comprehensive understanding of the etiology and pathogenesis of AAV and ultimately for improving the diagnosis, treatment, and prognosis of these patients.

DECLARATIONS

Authors’ contributions

Wrote and reviewed the biomarkers: Almeida S

Wrote and reviewed the pathogenesis: Neves MP

Wrote and reviewed the introduction: Da Silva Domingues V

All authors contributed for the final version of the paper.

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declare that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2021.

REFERENCES

1. Davies DJ, Moran JE, Niall JF, Ryan GB. Segmental necrotising glomerulonephritis with antineutrophil antibody: possible arbovirus aetiology? Br Med J (Clin Res Ed) 1982;285:606.

2. Kitching AR, Anders HJ, Basu N, et al. ANCA-associated vasculitis. Nat Rev Dis Primers 2020;6:71.

3. Seeliger B, Sznajd J, Robson JC, et al. Are the 1990 American College of Rheumatology vasculitis classification criteria still valid? Rheumatology (Oxford) 2017;56:1154-61.

4. Jennette JC, Falk RJ, Bacon PA, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum 2013;65:1-11.

5. Pearce FA, Craven A, Merkel PA, Luqmani RA, Watts RA. Global ethnic and geographic differences in the clinical presentations of anti-neutrophil cytoplasm antibody-associated vasculitis. Rheumatology (Oxford) 2017;56:1962-9.

6. Furuta S, Chaudhry AN, Arimura Y, et al. Comparison of the Phenotype and Outcome of Granulomatosis with Polyangiitis Between UK and Japanese Cohorts. J Rheumatol 2017;44:216-22.

7. 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;13:683-92.

8. Puéchal X, Pagnoux C, Perrodeau É, et al. French Vasculitis Study Group. Long-term outcomes among participants in the WEGENT trial of remission-maintenance therapy for granulomatosis with polyangiitis (Wegener’s) or microscopic polyangiitis. Arthritis Rheumatol 2016;68:690-701.

9. Yates M, Watts RA, Bajema IM, et al. EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis. Ann Rheum Dis 2016;75:1583-94.

10. Tanna A, Salama AD, Brookes P, Pusey CD. Familial granulomatosis with polyangiitis: three cases of this rare disorder in one Indoasian family carrying an identical HLA DPB1 allele. BMJ Case Rep 2012;2012:bcr0120125502.

11. Lyons PA, Rayner TF, Trivedi S, et al. Genetically distinct subsets within ANCA-associated vasculitis. N Engl J Med 2012;367:214-23.

12. Kawasaki A, Hasebe N, Hidaka M, et al. Protective role of HLA-DRB1*13:02 against microscopic polyangiitis and MPO-ANCA-positive vasculitides in a Japanese Population: a Case-Control Study. PLoS One 2016;11:e0154393.

13. Jones BE, Yang J, Muthigi A, et al. Gene-specific DNA methylation changes predict remission in patients with ANCA-associated vasculitis. J Am Soc Nephrol 2017;28:1175-87.

14. Ciavatta DJ, Yang J, Preston GA, et al. Epigenetic basis for aberrant upregulation of autoantigen genes in humans with ANCA vasculitis. J Clin Invest 2010;120:3209-19.

15. Nakazawa D, Masuda S, Tomaru U, Ishizu A. Pathogenesis and therapeutic interventions for ANCA-associated vasculitis. Nat Rev Rheumatol 2019;15:91-101.

16. Laudien M, Gadola SD, Podschun R, et al. Nasal carriage of Staphylococcus aureus and endonasal activity in Wegener’s granulomatosis as compared to rheumatoid arthritis and chronic Rhinosinusitis with nasal polyps. Clin Exp Rheumatol 2010;28:51-5.

17. Scott J, Hartnett J, Mockler D, Little MA. Environmental risk factors associated with ANCA associated vasculitis: a systematic mapping review. Autoimmun Rev 2020;19:102660.

18. Watts RA, Gonzalez-Gay MA, Lane SE, Garcia-Porrua C, Bentham G, Scott DG. Geoepidemiology of systemic vasculitis: comparison of the incidence in two regions of Europe. Ann Rheum Dis 2001;60:170-2.

19. Pearce FA, Lanyon PC, Watts RA, Grainge MJ, Abhishek A, Hubbard RB. Novel insights into the aetiology of granulomatosis with polyangiitis-a case-control study using the Clinical Practice Research Datalink. Rheumatology (Oxford) 2018;57:1002-10.

20. Gómez-Puerta JA, Gedmintas L, Costenbader KH. The association between silica exposure and development of ANCA-associated vasculitis: systematic review and meta-analysis. Autoimmun Rev 2013;12:1129-35.

21. Lane SE, Watts RA, Bentham G, Innes NJ, Scott DG. Are environmental factors important in primary systemic vasculitis? Arthritis Rheum 2003;48:814-23.

22. Stamp LK, Chapman PT, Francis J, et al. Association between environmental exposures and granulomatosis with polyangiitis in Canterbury, New Zealand. Arthritis Res Ther 2015;17:333.

23. Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science 2004;303:1532-5.

24. Sangaletti S, Tripodo C, Chiodoni C, et al. Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells toward ANCA induction and associated autoimmunity. Blood 2012;120:3007-18.

25. Hakkim A, Fürnrohr BG, Amann K, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 2010;107:9813-8.

26. Nakazawa D, Shida H, Tomaru U, et al. Enhanced formation and disordered regulation of NETs in myeloperoxidase-ANCA-associated microscopic polyangiitis. J Am Soc Nephrol 2014;25:990-7.

27. Hiwa R, Ohmura K, Arase N, et al. Myeloperoxidase/HLA Class II complexes recognized by autoantibodies in microscopic polyangiitis. Arthritis Rheumatol 2017;69:2069-80.

28. Leacy E, Brady G, Little MA. Pathogenesis of ANCA-associated vasculitis: an emerging role for immunometabolism. Rheumatology (Oxford) 2020;59:iii33-41.

29. Kronbichler A, Lee KH, Denicolò S, et al. Immunopathogenesis of ANCA-associated vasculitis. Int J Mol Sci 2020;21:7319.

30. Xiao H, Schreiber A, Heeringa P, Falk RJ, Jennette JC. Alternative complement pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic autoantibodies. Am J Pathol 2007;170:52-64.

31. Schreiber A, Xiao H, Jennette JC, Schneider W, Luft FC, Kettritz R. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am Soc Nephrol 2009;20:289-98.

32. Nakazawa D, Shida H, Kusunoki Y, et al. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun 2016;67:19-28.

33. Chen M, Kallenberg CG. ANCA-associated vasculitides--advances in pathogenesis and treatment. Nat Rev Rheumatol 2010;6:653-64.

34. Stegeman C, Tervaert J, de Jong P. Trimethoprim-sulfamethoxazole (co-trimoxazole) for the prevention of relapses of Wegener’s granulomatosis. Am J Ophthalmol 1996;122:606.

35. Polzer K, Karonitsch T, Neumann T, et al. Eotaxin-3 is involved in Churg-Strauss syndrome--a serum marker closely correlating with disease activity. Rheumatology (Oxford) 2008;47:804-8.

36. Csernok E, Hellmich B. Usefulness of vasculitis biomarkers in the era of the personalized medicine. Autoimmun Rev 2020;19:102514.

37. Brogan P, Eleftheriou D. Vasculitis update: pathogenesis and biomarkers. Pediatr Nephrol 2018;33:187-98.

38. Roth AJ, Ooi JD, Hess JJ, et al. Epitope specificity determines pathogenicity and detectability in ANCA-associated vasculitis. J Clin Invest 2013;123:1773-83.

39. Lally L, Spiera RF. Biomarkers in ANCA-associated vasculitis. Curr Rheumatol Rep 2013;15:363.

40. Morris AD, Rowbottom AW, Martin FL, Woywodt A, Dhaygude AP. Biomarkers in ANCA-associated vasculitis: potential pitfalls and future prospects. Kidney360 2021;2:586-97.

41. Domingues VM, Machado B, Santos J. ANCA-positive vasculitis: clinical implications of ANCA types and titers. Rev Assoc Med Bras (1992) 2016;62:434-40.

42. Suwanchote S, Rachayon M, Rodsaward P, et al. Anti-neutrophil cytoplasmic antibodies and their clinical significance. Clin Rheumatol 2018;37:875-84.

43. FijoŁek J, Wiatr E. Antineutrophil cytoplasmic antibodies (ANCA) - their role in pathogenesis, diagnosis, and treatment monitoring of ANCA-associated vasculitis. Cent Eur J Immunol 2020;45:218-27.

44. Gou SJ, Xu PC, Chen M, Zhao MH. Epitope analysis of anti-myeloperoxidase antibodies in patients with ANCA-associated vasculitis. PLoS One 2013;8:e60530.

45. Renn JH, Yang NP, Chueh CM, Lin CY, Lan TH, Chou P. Bone mass in schizophrenia and normal populations across different decades of life. BMC Musculoskelet Disord 2009;10:1.

46. Fussner LA, Hummel AM, Schroeder DR, et al. Rituximab in ANCA-Associated Vasculitis-Immune Tolerance Network Research Group. Factors determining the clinical utility of serial measurements of antineutrophil cytoplasmic antibodies targeting proteinase 3. Arthritis Rheumatol 2016;68:1700-10.

47. Schönermarck U, Lamprecht P, Csernok E, Gross WL. Prevalence and spectrum of rheumatic diseases associated with proteinase 3-antineutrophil cytoplasmic antibodies (ANCA) and myeloperoxidase-ANCA. Rheumatology (Oxford) 2001;40:178-84.

48. Mahr A, Katsahian S, Varet H, 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;72:1003-10.

49. Lionaki S, Blyth ER, Hogan SL, et al. Classification of antineutrophil cytoplasmic autoantibody vasculitides: the role of antineutrophil cytoplasmic autoantibody specificity for myeloperoxidase or proteinase 3 in disease recognition and prognosis. Arthritis Rheum 2012;64:3452-62.

50. Pepper RJ, Draibe JB, Caplin B, et al. RAVE-Immune Tolerance Network Research Group. Association of serum calprotectin (S100A8/A9) level with disease relapse in proteinase 3-antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheumatol 2017;69:185-93.

51. Kessenbrock K, Krumbholz M, Schönermarck U, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med 2009;15:623-5.

52. Manojlovic M, Juto A, Jonasdottir A, et al. Microparticles expressing myeloperoxidase as potential biomarkers in anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV). J Mol Med (Berl) 2020;98:1279-86.

53. Lange C, Csernok E, Moosig F, Holle JU. Immune stimulatory effects of neutrophil extracellular traps in granulomatosis with polyangiitis. Clin Exp Rheumatol 2017;35 Suppl 103:33-9.

54. Barnado A, Crofford LJ, Oates JC. At the bedside: neutrophil extracellular traps (NETs) as targets for biomarkers and therapies in autoimmune diseases. J Leukoc Biol 2016;99:265-78.

55. Lieberthal JG, Cuthbertson D, Carette S, et al. Vasculitis Clinical Research Consortium. urinary biomarkers in relapsing antineutrophil cytoplasmic antibody-associated vasculitis. J Rheumatol 2013;40:674-83.

56. Tam FW, Sanders JS, George A, et al. Urinary monocyte chemoattractant protein-1 (MCP-1) is a marker of active renal vasculitis. Nephrol Dial Transplant 2004;19:2761-8.

57. Ohlsson S, Bakoush O, Tencer J, Torffvit O, Segelmark M. Monocyte chemoattractant protein 1 is a prognostic marker in ANCA-associated small vessel vasculitis. Mediators Inflamm 2009;2009:584916.

58. O’Reilly VP, Wong L, Kennedy C, et al. Urinary soluble CD163 in active renal vasculitis. J Am Soc Nephrol 2016;27:2906-16.

59. Nagai M, Hirayama K, Ebihara I, Higuchi T, Shimohata H, Kobayashi M. Serum levels of the soluble haemoglobin scavenger receptor CD163 in MPO-ANCA-associated renal vasculitis. Scand J Rheumatol 2016;45:397-403.

60. Monach PA, Warner RL, Tomasson G, et al. Serum proteins reflecting inflammation, injury and repair as biomarkers of disease activity in ANCA-associated vasculitis. Ann Rheum Dis 2013;72:1342-50.

61. Dallos T, Heiland GR, Strehl J, et al. CCL17/thymus and activation-related chemokine in Churg-Strauss syndrome. Arthritis Rheum 2010;62:3496-503.

62. Vaglio A, Strehl JD, Manger B, et al. IgG4 immune response in Churg-Strauss syndrome. Ann Rheum Dis 2012;71:390-3.

63. Pepper RJ, Hamour S, Chavele KM, et al. Leukocyte and serum S100A8/S100A9 expression reflects disease activity in ANCA-associated vasculitis and glomerulonephritis. Kidney Int 2013;83:1150-8.

64. Szczeklik W, Sanak M, Mastalerz L, et al. 12-hydroxy-eicosatetraenoic acid (12-HETE): a biomarker of Churg-Strauss syndrome. Clin Exp Allergy 2012;42:513-22.

65. Henes FO, Chen Y, Bley TA, et al. Correlation of serum level of high mobility group box 1 with the burden of granulomatous inflammation in granulomatosis with polyangiitis (Wegener’s). Ann Rheum Dis 2011;70:1926-9.

66. Brogan PA, Dillon MJ. Endothelial microparticles and the diagnosis of the vasculitides. Intern Med 2004;43:1115-9.

67. Clarke LA, Hong Y, Eleftheriou D, et al. Endothelial injury and repair in systemic vasculitis of the young. Arthritis Rheum 2010;62:1770-80.

68. Erdbruegger U, Grossheim M, Hertel B, et al. Diagnostic role of endothelial microparticles in vasculitis. Rheumatology (Oxford) 2008;47:1820-5.

69. Eleftheriou D, Hong Y, Klein NJ, Brogan PA. Thromboembolic disease in systemic vasculitis is associated with enhanced microparticle-mediated thrombin generation. J Thromb Haemost 2011;9:1864-7.

70. Eleftheriou D, Ganesan V, Hong Y, Klein NJ, Brogan PA. Endothelial injury in childhood stroke with cerebral arteriopathy: a cross-sectional study. Neurology 2012;79:2089-96.

71. Woywodt A, Streiber F, de Groot K, Regelsberger H, Haller H, Haubitz M. Circulating endothelial cells as markers for ANCA-associated small-vessel vasculitis. Lancet 2003;361:206-10.

72. Závada J, Kideryová L, Pytlík R, Hrusková Z, Tesar V. Reduced number of endothelial progenitor cells is predictive of early relapse in anti-neutrophil cytoplasmic antibody-associated vasculitis. Rheumatology (Oxford) 2009;48:1197-201.

73. Cantin L, Chartrand-Lefebvre C, Marcotte F, Pressacco J, Ducharme A, Lapierre C. Coronary artery noninvasive imaging in adult Kawasaki disease. Clin Imaging 2009;33:181-7.

74. van Dam LS, Dirikgil E, Bredewold EW, et al. Proteinase-3-anti-neutrophil cytoplasmic antibodies (PR3-ANCAs) predict relapses in ANCA-associated vasculitis patients after rituximab. Nephrol Dial Transplant 2020:gfaa066.

75. Smith RM, Jones RB, Guerry MJ, et al. Rituximab for remission maintenance in relapsing antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum 2012;64:3760-9.

76. Bunch DO, McGregor JG, Khandoobhai NB, et al. Decreased CD5+ B cells in active ANCA vasculitis and relapse after rituximab. Clin J Am Soc Nephrol 2013;8:382-91.

77. Unizony S, Lim N, Phippard DJ, et al. Peripheral CD5+ B cells in antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheumatol 2015;67:535-44.

78. Bader L, Koldingsnes W, Nossent J. B-lymphocyte activating factor levels are increased in patients with Wegener’s granulomatosis and inversely correlated with ANCA titer. Clin Rheumatol 2010;29:1031-5.

79. Dolff S, Witzke O, Wilde B. Th17 cells in renal inflammation and autoimmunity. Autoimmun Rev 2019;18:129-36.

80. von Borstel A, Lintermans LL, Heeringa P, et al. Circulating CD24hiCD38hi regulatory B cells correlate inversely with the ThEM17 cell frequency in granulomatosis with polyangiitis patients. Rheumatology (Oxford) 2019; doi: 10.1093/rheumatology/key412.

81. Guillevin L, Pagnoux C, Karras A, et al. Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis. A prospective study in 117 patients. La Presse Médicale 2013;42:679.

82. Li K, Zhang Y, Zhang Y, et al. Tyrosine kinase Fyn promotes osteoarthritis by activating the β-catenin pathway. Ann Rheum Dis 2018;77:935-43.

83. Wang Y, Zhang S, Zhang N, et al. Reduced activated regulatory T cells and imbalance of Th17/activated Treg cells marks renal involvement in ANCA-associated vasculitis. Mol Immunol 2020;118:19-29.

84. Deng J, Wei Y, Fonseca VR, Graca L, Yu D. T follicular helper cells and T follicular regulatory cells in rheumatic diseases. Nat Rev Rheumatol 2019;15:475-90.

85. Gou SJ, Yuan J, Wang C, Zhao MH, Chen M. Alternative complement pathway activation products in urine and kidneys of patients with ANCA-associated GN. Clin J Am Soc Nephrol 2013;8:1884-91.

86. Tesar V, Hruskova Z. Avacopan in the treatment of ANCA-associated vasculitis. Expert Opin Investig Drugs 2018;27:491-6.

87. Yuan J, Gou SJ, Huang J, Hao J, Chen M, Zhao MH. C5a and its receptors in human anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis. Arthritis Res Ther 2012;14:R140.

88. Gou SJ, Yuan J, Chen M, Yu F, Zhao MH. Circulating complement activation in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Kidney Int 2013;83:129-37.

89. Jayne DRW, Merkel PA, Schall TJ, Bekker P. ADVOCATE Study Group. Avacopan for the Treatment of ANCA-Associated Vasculitis. N Engl J Med 2021;384:599-609.

90. Manenti L, Vaglio A, Gnappi E, et al. Association of serum C3 concentration and histologic signs of thrombotic microangiopathy with outcomes among patients with ANCA-associated renal vasculitis. Clin J Am Soc Nephrol 2015;10:2143-51.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Almeida S, Neves PM, Da Silva Domingues V. Pathogenesis and biomarkers in ANCA-associated vasculitis. Vessel Plus 2021;5:44. http://dx.doi.org/10.20517/2574-1209.2021.66

AMA Style

Almeida S, Neves PM, Da Silva Domingues V. Pathogenesis and biomarkers in ANCA-associated vasculitis. Vessel Plus. 2021; 5: 44. http://dx.doi.org/10.20517/2574-1209.2021.66

Chicago/Turabian Style

Almeida, Sofia, Pedro Maia Neves, Vital Da Silva Domingues. 2021. "Pathogenesis and biomarkers in ANCA-associated vasculitis" Vessel Plus. 5: 44. http://dx.doi.org/10.20517/2574-1209.2021.66

ACS Style

Almeida, S.; Neves PM.; Da Silva Domingues V. Pathogenesis and biomarkers in ANCA-associated vasculitis. Vessel Plus. 2021, 5, 44. http://dx.doi.org/10.20517/2574-1209.2021.66

About This Article

Special Issue

© The Author(s) 2021. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
3481
Downloads
969
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 64 clicks
Like This Article 0 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Vessel Plus
ISSN 2574-1209 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/