MedPath

Avacopan Advanced Drug Monograph

Published:Jun 13, 2025

Generic Name

Avacopan

Brand Names

Tavneos

Drug Type

Small Molecule

Chemical Formula

C33H35F4N3O2

CAS Number

1346623-17-3

Associated Conditions

Severe, active Granulomatosis With Polyangiitis, Severe, active Microscopic Polyangiitis (MPA), Microscopic Polyangiitis (MPA), Severe Anti-neutrophil cytoplasmic antibody positive vasculitis, Granulomatosis With Polyangiitis

Avacopan (Tavneos): A Comprehensive Clinical and Pharmacological Review

1. Introduction to Avacopan

[1.1. Overview and Therapeutic Class]

Avacopan, marketed under the trade name Tavneos, is an orally bioavailable small molecule medication.[1] It functions as a selective, allosteric antagonist of the human complement 5a receptor (C5aR, also known as C5aR1 or CD88).[1] This mechanism of action positions it as a targeted immunomodulatory agent. Both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have designated Avacopan as a first-in-class medication.[1] This designation is significant as it underscores the novelty of Avacopan's therapeutic approach to ANCA-associated vasculitis (AAV). As a first-in-class agent, Avacopan targets a pathway in a manner distinct from previously available treatments, offering potential advantages for patients, particularly in addressing unmet medical needs. However, the novelty also implies that clinicians and researchers will continue to accumulate long-term real-world data to fully understand its optimal use, unique aspects of its side effect profile, and its long-term impact compared to more established therapeutic strategies.

[1.2. Approved Indications]

Avacopan is indicated as an adjunctive treatment for adult patients with severe active anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV), specifically targeting two main forms: granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA).[1] It is designed to be used in combination with standard therapeutic regimens, which typically include glucocorticoids along with either rituximab or cyclophosphamide.[1] It is important to recognize that Avacopan's role is adjunctive; it is added to an existing comprehensive treatment strategy. While a primary goal of Avacopan therapy is to reduce the substantial burden of glucocorticoid use and its associated toxicities, it does not entirely eliminate the need for glucocorticoids in all approved regimens, particularly as per the FDA indication in the United States.[3] The overarching aim is to enhance the overall efficacy of the treatment regimen and/or mitigate the toxicity of its other components, most notably glucocorticoids.

[1.3. Brief Background on ANCA-Associated Vasculitis (AAV)]

ANCA-associated vasculitis encompasses a group of rare, severe, and systemic autoimmune diseases. These conditions are characterized by necrotizing inflammation of small blood vessels and are typified by the presence of anti-neutrophil cytoplasmic autoantibodies (ANCAs) in the patient's serum.[3] The estimated incidence of AAV is low, approximately 3 cases per 100,000 individuals per year, highlighting its rarity.[3] The AAV spectrum includes granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), eosinophilic granulomatosis with polyangiitis (EGPA), and drug-induced AAV; Avacopan's current approvals are primarily for GPA and MPA.[3]

AAV can lead to severe and life-threatening organ damage, with a particular predilection for the kidneys, where it can cause necrotizing and crescentic glomerulonephritis (NCGN), and the respiratory tract.[3] The pathophysiology of AAV is complex but centrally involves the ANCA-mediated activation of neutrophils. These activated neutrophils adhere to and damage endothelial cells lining the small blood vessels. A critical component in this inflammatory cascade is the complement system, specifically the C5a-C5aR axis. The anaphylatoxin C5a, generated via the alternative complement pathway, binds to its receptor (C5aR) on neutrophils, further amplifying inflammation by promoting neutrophil chemotaxis, activation, and degranulation.[3]

The combination of AAV's rarity, its potential for severe organ damage, and the significant toxicities associated with traditional treatments—especially high-dose and long-term glucocorticoid therapy—creates a substantial unmet medical need. This context underscores the importance of developing targeted therapies like Avacopan, which aim to improve clinical outcomes while reducing the treatment-related burden for patients.

2. Chemical and Physical Properties

[2.1. Chemical Name and Structure]

Avacopan was formerly known by the research code CCX168.[1] One of its formal chemical names is (2R,3S)-2-[4-(cyclopentylamino)phenyl]-1-(2-fluoro-6-methyl-benzoyl)piperidine-3-carboxylate, though other systematic names exist.[12] Two-dimensional structural representations are available in various chemical databases.

[2.2. Molecular Formula, Molecular Weight, CAS Number]

The key chemical identifiers for Avacopan are:

  • Molecular Formula: C33​H35​F4​N3​O2​ [1]
  • Molar Mass / Molecular Weight: Approximately 581.65 g/mol (values range from 581.64 to 581.656 g·mol⁻¹ depending on the source) [1]
  • CAS Number: 1346623-17-3 [1]

Other notable identifiers include:

  • DrugBank ID: DB15011 [1]
  • PubChem CID: 49841217 [1]
  • UNII: O880NM097T [1]
  • KEGG: D11093 [1]
  • ChEMBL: ChEMBL3989871 [1]

The consistency of these identifiers across multiple international databases provides unambiguous confirmation of the specific chemical entity. Avacopan's classification as a "small molecule" [4] is consistent with its chemical properties, oral bioavailability, and its susceptibility to cytochrome P450-mediated metabolism, which are characteristic features discussed in its pharmacokinetic profile.

3. Pharmacology

[3.1. Mechanism of Action]

Avacopan exerts its therapeutic effect by acting as a selective antagonist of the complement 5a receptor (C5aR), also identified as C5aR1 or CD88.[1] It functions as an allosteric antagonist, meaning it binds to a site on the receptor distinct from the C5a binding site, thereby modulating receptor activity.[3]

The interaction between the anaphylatoxin C5a and its receptor C5aR is a pivotal step in the pro-inflammatory cascade that drives ANCA-associated vasculitis.[3] C5a, a potent inflammatory mediator generated through activation of the alternative complement pathway, binds to C5aR expressed on the surface of neutrophils and other immune cells. This ligand-receptor binding triggers a series of downstream events critical to the pathology of AAV, including neutrophil activation, chemotaxis (directed migration to sites of inflammation), adhesion to the vascular endothelium, degranulation (release of cytotoxic enzymes and inflammatory mediators), and the generation of reactive oxygen species.[4] These processes collectively contribute to the necrotizing inflammation and damage of small blood vessels.

By specifically blocking the C5aR, Avacopan inhibits C5a-mediated neutrophil activation and migration, thereby attenuating this amplification loop of inflammation.[4] Avacopan's mechanism differs fundamentally from that of C5 complement protein inhibitors (such as eculizumab and ravulizumab), which are biologic agents that target the C5 protein itself, preventing its cleavage into C5a and C5b. In contrast, Avacopan is a small molecule that targets the C5a receptor directly.[4] The potency of Avacopan is high, as indicated by its low half-maximal inhibitory concentration (

IC50​) values for C5aR, reported in the range of 0.1 nM to 0.2 nM in various in vitro assays.[6]

The targeted nature of Avacopan's action represents a significant refinement compared to the broad immunosuppression afforded by traditional therapies like glucocorticoids. Glucocorticoids exert widespread systemic effects, leading to a well-documented array of adverse effects. By selectively inhibiting a key pro-inflammatory mediator (C5a) at its specific receptor on neutrophils, Avacopan aims to achieve a more precise modulation of the immune response. This targeted approach holds the potential to reduce inflammation effectively with fewer off-target effects, thereby improving the therapeutic index. The allosteric nature of its antagonism might also confer advantages in terms of receptor modulation compared to competitive antagonists that directly compete with the natural ligand.

[3.2. Pharmacodynamics]

The pharmacodynamic effects of Avacopan provide evidence of its target engagement and functional inhibition of key neutrophil activities implicated in the pathogenesis of AAV. In vitro studies have demonstrated that Avacopan effectively blocks several C5a-mediated neutrophil responses, including C5a-induced intracellular calcium mobilization, upregulation of CD11b (an adhesion molecule crucial for neutrophil trafficking and activation), and the respiratory burst (production of reactive oxygen species).[6]

Clinical pharmacodynamic studies in healthy volunteers have shown that an oral dose of Avacopan 30 mg twice daily results in approximately 94% blockade of C5aR, as measured by the inhibition of C5a-induced CD11b upregulation on neutrophils.[11] This high level of receptor occupancy at clinically relevant doses supports the pharmacological rationale for its efficacy in AAV.

While the primary mechanism of C5aR blockade is well-established, some sources note that the exact downstream mechanisms contributing to its full therapeutic effect in the complex in vivo milieu of ANCA-associated vasculitis are "not well understood".[15] This likely refers to the complete spectrum of downstream immunological consequences beyond the direct effects on neutrophil C5aR signaling, given the multifaceted nature of AAV. Nevertheless, the documented pharmacodynamic effects provide a clear link between Avacopan's molecular action (C5aR antagonism) and its intended biological effect on the key pathogenic cells involved in AAV.

[3.3. Pharmacokinetics]

The pharmacokinetic profile of Avacopan has been characterized in several studies, providing insights into its absorption, distribution, metabolism, and excretion (ADME) properties.

Absorption:

Avacopan is orally bioavailable.3 It is recommended to be administered with food.2 The presence of food significantly influences Avacopan's absorption; administration with a high-fat, high-calorie meal was found to increase the peak plasma concentration (

Cmax​) by approximately 8% and the area under the plasma concentration-time curve (AUC) by a substantial 72%. Food also delays the time to reach Cmax​ (Tmax​) by about 4 hours.[15] When administered without food,

Tmax​ is typically achieved approximately 2 hours post-dose.[15] The marked increase in AUC with food underscores the importance of administering Avacopan with meals to ensure consistent and optimal drug exposure, which is critical for achieving predictable therapeutic effects. This is a key counseling point for patients to maintain adherence and efficacy.

Distribution:

Avacopan is extensively bound to plasma proteins, with a binding percentage reported as >99.9%.15 Such high plasma protein binding implies that only a very small fraction of the drug in circulation is unbound and therefore pharmacologically active. While changes in protein binding due to other drugs or altered physiological states (e.g., hypoalbuminemia) could theoretically affect the free fraction of highly bound drugs, this is generally less of a concern unless the displacing agent is also highly bound and present at high concentrations. Information regarding Avacopan's distribution into human milk is currently unavailable.2

Metabolism:

Avacopan is principally metabolized by the cytochrome P450 3A4 (CYP3A4) enzyme system in the liver.9 Notably, Avacopan itself also acts as an inhibitor of CYP3A4.1 It does not significantly inhibit other major CYP isoforms such as CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP2D6 at clinically relevant concentrations.15

Avacopan is metabolized to an active metabolite, designated M1.15 This M1 metabolite also possesses pharmacological activity and has its own metabolic profile; it may inhibit CYP2C9 and CYP3A4, and potentially induce CYP3A4, CYP1A2, and CYP2B6.15 Furthermore, the M1 metabolite is a substrate of the efflux transporter P-glycoprotein (P-gp).15

The dual role of Avacopan as both a substrate and an inhibitor of CYP3A4, coupled with the activity of its M1 metabolite, is of critical clinical importance. This profile creates a high potential for drug-drug interactions (DDIs). Co-administered medications that are strong inducers or inhibitors of CYP3A4 can significantly alter Avacopan plasma concentrations, thereby affecting its efficacy or safety. Conversely, Avacopan can influence the plasma concentrations of other drugs that are substrates of CYP3A4. The M1 metabolite's own inhibitory and inductive potential adds a further layer of complexity to predicting the full spectrum of DDIs.

Excretion:

Specific details regarding the quantitative routes of excretion (e.g., percentages excreted renally versus facally) are not extensively detailed in the provided information, though hepatic metabolism via CYP3A4 is established as the principal pathway for its elimination.

Steady-State Concentrations and Accumulation:

Following repeated oral administration, steady-state plasma concentrations of Avacopan are achieved in approximately 13 weeks.15 The drug exhibits an accumulation of approximately fourfold at steady state compared to single-dose exposure.15 The relatively long time to reach steady state (13 weeks) implies that the full therapeutic effect, as well as the full extent of any concentration-dependent adverse effects, might not become apparent for several months after initiating therapy or adjusting the dose. This extended timeframe necessitates patience in evaluating treatment response and caution against overly frequent dose modifications. The fourfold accumulation is a significant factor that is considered in the development of the dosing regimen to ensure that steady-state concentrations remain within the therapeutic and safe range.

Table 1: Key Pharmacokinetic Parameters of Avacopan

ParameterValue / DescriptionSource(s)
AdministrationOral, with food5
Tmax​ (without food)~2 hours15
Food Effect (high-fat meal)Cmax​ ↑ by 8%, AUC ↑ by 72%, Tmax​ delayed by ~4 hours15
Plasma Protein Binding>99.9%15
Primary Metabolizing EnzymeCYP3A49
Avacopan as CYP InhibitorInhibits CYP3A41
Active MetaboliteM1 (may inhibit CYP2C9, CYP3A4; may induce CYP3A4, CYP1A2, CYP2B6; P-gp substrate)15
Time to Steady State~13 weeks15
Accumulation Factor~Fourfold15

4. Clinical Efficacy in ANCA-Associated Vasculitis

[4.1. Overview of Clinical Development Program]

The clinical development of Avacopan (formerly CCX168) for AAV followed a structured path, progressing through Phase 1 studies in healthy volunteers, Phase 2 proof-of-concept and dose-ranging studies (CLEAR and CLASSIC), and culminating in a pivotal Phase 3 trial (ADVOCATE).[4]

The Phase 2 studies, CLEAR and CLASSIC, were instrumental in exploring various doses of Avacopan and its potential as a glucocorticoid-sparing agent. The CLEAR trial (published 2017) randomized 67 AAV patients to one of three arms: standard prednisone taper plus Avacopan placebo, a reduced prednisone taper (20 mg initial) plus Avacopan 30 mg twice daily, or Avacopan 30 mg twice daily plus prednisone placebo. All patients received standard induction immunosuppression (rituximab or cyclophosphamide). The study demonstrated that Avacopan 30 mg twice daily (with prednisone placebo) was non-inferior to a standard 60 mg prednisone taper in terms of Birmingham Vasculitis Activity Score (BVAS) response (≥50% reduction) at week 12.[11] The CLASSIC trial further evaluated Avacopan (10 mg or 30 mg twice daily) or placebo, all in conjunction with a standard prednisone taper and background immunosuppression. This study also showed non-inferiority for BVAS response at week 12 for the Avacopan groups compared to placebo.[4]

Collectively, these Phase 2 trials provided crucial data supporting the 30 mg twice-daily dose of Avacopan and indicated that it could potentially replace or significantly reduce high-dose glucocorticoid regimens without compromising short-term disease activity control. This systematic, phased approach, demonstrating a consistent signal of non-inferiority in early disease activity reduction, laid a strong foundation and provided the rationale for proceeding to the larger, more definitive ADVOCATE Phase 3 trial.

[4.2. The ADVOCATE Trial (Phase 3 - NCT02994927)]

The ADVOCATE trial was a cornerstone in establishing the efficacy and safety of Avacopan for AAV and formed the primary basis for its regulatory approvals.

Trial Design:

ADVOCATE was a 52-week, multinational, multicenter, randomized, double-blind, double-dummy, active-controlled trial.1 A total of 331 adult patients with severe, active GPA or MPA were randomized in a 1:1 ratio to one of two treatment groups 1:

  1. Avacopan group (N=166): Received Avacopan 30 mg orally twice daily plus a prednisone-matching placebo for 20 weeks.
  2. Prednisone group (N=165, with 164 analyzed in some reports): Received a standard oral prednisone taper (starting at 60 mg/day and tapered to 0 mg/day over 20 weeks) plus an Avacopan-matching placebo twice daily.[19]

All patients in both groups also received standard background immunosuppressive therapy, which consisted of either intravenous rituximab (375 mg/m² weekly for 4 weeks) or cyclophosphamide (administered intravenously or orally for 13-14 weeks, followed by oral azathioprine for maintenance).[4]

Patient Population:

Eligible participants were adults with newly diagnosed or relapsing GPA or MPA, a positive ANCA test (anti-PR3 or anti-MPO), and an estimated glomerular filtration rate (eGFR) of at least 15 mL/min/1.73 m².18 The mean BVAS at baseline was approximately 16, indicating substantial disease activity.19

Primary Endpoints:

The trial had two primary efficacy endpoints 1:

  1. Remission at Week 26: Defined as a BVAS of 0 and no glucocorticoid use for AAV in the 4 weeks preceding week 26. This endpoint was tested for non-inferiority of Avacopan to prednisone taper (with a pre-specified non-inferiority margin of 20 percentage points) and subsequently for superiority.
  2. Sustained Remission at Week 52: Defined as achieving remission at week 26 and maintaining remission at week 52, with no relapse occurring between weeks 26 and 52, and no glucocorticoid use for AAV in the 4 weeks preceding week 52. This endpoint was also tested for non-inferiority and then for superiority.

Efficacy Results vs. Prednisone Taper:

  • Remission at Week 26: The Avacopan group demonstrated a remission rate of 72.3%, compared to 70.1% in the prednisone group.[4] Avacopan met the criterion for non-inferiority to prednisone taper for this endpoint (estimated common difference: 3.4 percentage points; 95% CI, -6.0 to 12.8; p<0.001 for non-inferiority).[19] However, Avacopan did not demonstrate superiority over prednisone taper for remission at week 26 (p=0.24).[19]
  • Sustained Remission at Week 52: For this endpoint, 65.7% of patients in the Avacopan group achieved sustained remission, compared to 54.9% in the prednisone group.[4] Avacopan was found to be non-inferior to prednisone taper (p<0.001) and, importantly, also demonstrated superiority (estimated common difference: 12.5 percentage points; 95% CI, 2.6 to 22.3; p=0.007 or p=0.013, depending on the source analysis).[4]

Glucocorticoid-Sparing Effect and Reduction in Glucocorticoid Toxicity:

A key finding of the ADVOCATE trial was the significant glucocorticoid-sparing effect of Avacopan. Patients in the Avacopan group received a substantially lower cumulative dose of glucocorticoids over the 52-week study period (mean dose approximately 1676 mg to 1731 mg prednisone equivalent) compared to those in the prednisone taper group (mean dose approximately 3687 mg to 3847 mg prednisone equivalent).11 This reduction in glucocorticoid exposure translated into clinically meaningful benefits, as evidenced by lower glucocorticoid-related toxicity in the Avacopan group, measured using the Glucocorticoid Toxicity Index (GTI).11 Furthermore, patients treated with Avacopan reported improvements in health-related quality of life (HRQoL) measures.11

Impact on Renal Outcomes:

The ADVOCATE trial also yielded positive signals regarding renal outcomes. Patients in the Avacopan group exhibited a greater improvement in eGFR at week 52 compared to the prednisone group.11 Additionally, a faster reduction in albuminuria, as measured by the urinary albumin-to-creatinine ratio (UACR), was observed in the Avacopan group.18 A post-hoc analysis suggested that the beneficial effect on eGFR was even more pronounced in patients who had a baseline eGFR below 20 mL/min/1.73 m².11

Subgroup Analyses:

  • Rituximab Subgroup: A pre-specified subgroup analysis focused on the 214 patients (64.8% of the trial population) who received rituximab as their background immunosuppressive therapy. In this subgroup, Avacopan treatment resulted in a remission rate at week 26 of 77.6% versus 75.7% for the prednisone taper group. For sustained remission at week 52, the rates were 71.0% for Avacopan versus 56.1% for prednisone taper. The relapse rate after achieving remission at any time was notably lower in the Avacopan plus rituximab group (8.7%) compared to the prednisone taper plus rituximab group (20.2%).[18]
  • The superiority of Avacopan for sustained remission was reportedly more pronounced in patients with MPA, those who were MPO-ANCA positive, and those with relapsing disease at baseline.[11]
  • Glucocorticoid-Free Subgroup: An interesting post-hoc analysis examined a small subgroup of 28 patients in the Avacopan arm who did not receive any protocol-defined or non-study-supplied glucocorticoids during the first 29 days of the trial. Within this group, 11 patients received no glucocorticoids at any point during the 52-week study. Remarkably, all 11 of these patients achieved remission at week 26 and sustained remission at week 52.[22]

The ADVOCATE trial's findings are pivotal. While Avacopan did not demonstrate superiority for inducing remission at the 26-week mark, its clear superiority in sustaining remission through 52 weeks is a highly significant outcome. This, combined with the substantial reduction in overall glucocorticoid exposure, diminished glucocorticoid-related toxicity, and favorable signals on renal function and quality of life, forms the core of Avacopan's clinical value proposition. These results suggest that Avacopan facilitates the maintenance of long-term disease control with a significantly reduced steroid burden, which has been a long-standing goal in AAV management. The data from the rituximab subgroup are particularly compelling and support its use with this standard background therapy. Although derived from a small, post-hoc analyzed subgroup, the outcomes in patients who received Avacopan without any glucocorticoids [22] hint at the exciting potential for even greater steroid avoidance in carefully selected AAV patients in the future, a prospect that warrants further investigation.

[4.3. Real-World Evidence (RWE)]

Following regulatory approvals, real-world evidence (RWE) on Avacopan's use in routine clinical practice is beginning to emerge. These studies are crucial as they often include patients with a broader range of disease severity and characteristics than those enrolled in pivotal clinical trials, such as individuals with very low eGFR (e.g., <15 mL/min/1.73 m²) or those with refractory disease, who might have been excluded from ADVOCATE.[11]

A multicenter observational cohort study from the UK, involving 61 AAV patients treated with Avacopan, reported notable differences from the ADVOCATE trial population. For instance, 18% of patients in this real-world cohort had an eGFR below 15 mL/min/1.73 m² at Avacopan initiation. Furthermore, Avacopan was often started later in the disease course, and it was more frequently combined with other immunosuppressants (beyond the standard rituximab or cyclophosphamide) than in the ADVOCATE trial.[23]

In this UK RWE cohort, a clinical response at 6 months (defined as BVAS version 3 score ≤3 and a prednisolone dose ≤5 mg/day) was achieved by 63% of patients. Importantly, 37% of patients were able to discontinue glucocorticoids entirely. Among patients with an initial eGFR <15 mL/min/1.73 m², 78% showed an improvement to an eGFR >15 mL/min/1.73 m² by 6 months.23

The rate of complete remission according to the strict ADVOCATE trial definition (BVAS=0 and no glucocorticoids for 4 weeks) was lower in this real-world setting (11.5% at 6 months). However, the investigators noted that this less frequent achievement of "complete remission" did not necessarily lead to an escalation of immunosuppressive therapy by clinicians, suggesting that other measures of clinical improvement might be guiding treatment decisions in practice.23 After applying inverse probability weighting to compare with a historical cohort, Avacopan-treated patients in this RWE study were found to be 2.9 times more likely to achieve the defined clinical response.23

Other RWE sources also support the use of Avacopan in patients with refractory disease and in those with severe rapidly progressive glomerulonephritis (RPGN) or diffuse alveolar hemorrhage (DAH), demonstrating good remission rates and renal recovery, sometimes exceeding the improvements observed in specific subgroups of the ADVOCATE trial.[11]

The emergence of RWE is vital for understanding how Avacopan performs across the heterogeneous spectrum of AAV patients encountered in daily clinical practice. These early real-world data generally suggest effectiveness, including in patients with more severe renal impairment than studied in ADVOCATE, and reinforce the glucocorticoid-sparing potential of Avacopan. The observation that lower rates of "complete remission" (by strict trial criteria) in real-world settings do not always prompt treatment intensification is an important nuance, highlighting potential differences between formal trial endpoints and the pragmatic clinical goals pursued in routine care.

Table 2: Summary of Key Efficacy Outcomes from the ADVOCATE Trial (Intent-to-Treat Population)

EndpointAvacopan Group (N=166) Result (%)Prednisone Taper Group (N=164/165) Result (%)Treatment Difference (95% CI)P-valueSource(s)
Remission at Week 2672.370.13.4% (-6.0 to 12.8)<0.001 (NI)a, 0.24 (Sup)b4
Sustained Remission at Week 5265.754.912.5% (2.6 to 22.3)<0.001 (NI)a, 0.007 (Sup)b4
Mean Cumulative GC Dose (mg prednisone equiv. over 52 weeks)~1676 - 1731~3847 - 3687Substantially lower in Avacopan groupSignificant11
Relapse Rate (RTX Subgroup)c8.7 (9/104)20.2 (21/104)HR 0.42 (0.19 to 0.91)-18
eGFR Improvement at Week 52 (mL/min/1.73m²)Greater increaseLess increaseLS Mean Diff. Favored Avacopan-18

NI: Non-inferiority; Sup: Superiority. aP-value for non-inferiority. bP-value for superiority. cRelapse rate after remission at any time in patients receiving rituximab as background therapy.

5. Safety and Tolerability

[5.1. Overview of Adverse Reactions]

The safety profile of Avacopan has been evaluated in clinical trials, primarily the ADVOCATE study, and is also being characterized through ongoing real-world experience.

Common Adverse Effects:

In the ADVOCATE trial, the most common adverse effects reported in ≥5% of patients and occurring at a higher incidence in the Avacopan group compared to the prednisone taper group included 1:

  • Nausea (e.g., 23.5% in Avacopan group vs. 20.7% in prednisone group)
  • Headache (e.g., 20.5% vs. 14.0%)
  • Hypertension (e.g., 18.1% vs. 17.7%)
  • Diarrhea (e.g., 15.1% vs. 14.6%)
  • Vomiting (e.g., 15.1% vs. 12.8%)
  • Rash (e.g., 11.4% vs. 7.9%)
  • Fatigue (e.g., 10.2% vs. 9.1%)
  • Upper abdominal pain (e.g., 6.6% vs. 6.1%)
  • Dizziness (e.g., 6.6% vs. 6.1%)
  • Blood creatinine increased (e.g., 6.0% vs. 4.9%)
  • Paresthesia (e.g., 5.4% vs. 4.3%)

Generic listings outside of specific trial data sometimes report higher overall percentages for some common side effects like nausea (39%) and headache (34%).[1]

Table 3: Common Adverse Reactions (Reported in ≥5% of Patients and Higher in the TAVNEOS® Arm vs Active Control Arm) in the ADVOCATE Phase 3 Trial [8]

Adverse ReactionTAVNEOS® (N=166), n (%)Active Control (Prednisone Taper) Arm (N=164), n (%)
Nausea39 (23.5)34 (20.7)
Headache34 (20.5)23 (14.0)
Hypertension30 (18.1)29 (17.7)
Diarrhea25 (15.1)24 (14.6)
Vomiting25 (15.1)21 (12.8)
Rash19 (11.4)13 (7.9)
Fatigue17 (10.2)15 (9.1)
Upper abdominal pain11 (6.6)10 (6.1)
Dizziness11 (6.6)10 (6.1)
Blood creatinine increased10 (6.0)8 (4.9)
Paresthesia9 (5.4)7 (4.3)

Serious Adverse Effects (SAEs):

In the ADVOCATE trial, the overall incidence of SAEs (excluding worsening vasculitis) was comparable between the treatment groups: 37.3% in patients receiving Avacopan and 39.0% in those receiving prednisone.19 In the rituximab subgroup of ADVOCATE, SAEs occurred in 34.6% of Avacopan-treated patients and 39.3% of prednisone-treated patients.18

Longer-term data from an Early Access Program (EAP), involving 59 patients treated for a median duration of 16 months, showed that 20.3% experienced at least one adverse event, and 6 of these 59 patients (10.2%) experienced a serious adverse event. The most common AEs in this longer-term cohort were gastrointestinal disorders and infections, with COVID-19 being frequently reported.24

While many of the common adverse events are gastrointestinal or general in nature (e.g., headache, fatigue), the similar overall SAE rates in ADVOCATE between the Avacopan and prednisone groups are noteworthy. This suggests that while Avacopan successfully reduces steroid-specific toxicities, it possesses its own distinct adverse event profile that clinicians must manage. The reported increase in blood creatinine requires careful clinical assessment to differentiate between potential drug-related effects and manifestations of the underlying AAV or its complications. The manageable safety profile observed in longer-term EAP data provides some reassurance for extended use, although this is in a less rigorously controlled setting than a clinical trial.

[5.2. Warnings and Precautions]

The use of Avacopan is associated with several important warnings and precautions that necessitate careful patient selection, monitoring, and management.

Hepatotoxicity:

Serious cases of hepatic injury, including life-threatening events and instances of clinically apparent liver injury with jaundice, have been observed in patients taking Avacopan.3 In clinical trials, elevations in serum aminotransferases (ALT or AST) occurred in approximately 12% of patients receiving Avacopan, although these were largely asymptomatic and transient.10 Serious hepatic adverse events involving aminotransferase elevations greater than 3 times the upper limit of normal (ULN) were reported in 5.4% of Avacopan-treated patients compared to 3.6% in the control group in pooled analyses.10

  • Monitoring: It is mandatory to obtain a liver test panel (including ALT, AST, alkaline phosphatase, and total bilirubin) before initiating Avacopan therapy. Monitoring should continue every 4 weeks for the first 6 months of treatment and as clinically indicated thereafter.[7]
  • Management: If ALT or AST levels rise to >3 times ULN, treatment should be promptly evaluated and pausing Avacopan considered. Avacopan must be discontinued if ALT or AST levels exceed 5 times ULN, or if they are >3 times ULN accompanied by a total bilirubin elevation >2 times ULN, until Avacopan-induced liver injury can be ruled out.[7]
  • Use in Liver Disease: Avacopan is not recommended for patients with active, untreated, and/or uncontrolled chronic liver disease (e.g., chronic active hepatitis B, untreated hepatitis C, uncontrolled autoimmune hepatitis) and cirrhosis.[7]

Serious Hypersensitivity Reactions:

Cases of angioedema, including at least one serious event requiring hospitalization, have occurred in clinical trials of Avacopan.7 If angioedema occurs, Avacopan must be discontinued immediately, and appropriate medical management instituted. Avacopan should not be readministered unless an alternative cause for the angioedema has been definitively established.7

Hepatitis B Virus (HBV) Reactivation:

Reactivation of HBV, including instances of life-threatening hepatitis B, has been observed during the Avacopan clinical program. Two cases of HBV reactivation were noted in pre-registration studies, although these patients were also receiving other therapies known to induce reactivation.7

  • Screening: All patients should be screened for HBV infection (including HBsAg and anti-HBc tests) before starting Avacopan.[7]
  • Management of Prior Infection: For patients with evidence of prior HBV infection, consultation with physicians experienced in HBV management is essential. These patients require monitoring during Avacopan therapy and for at least 6 months following its discontinuation.[7] If HBV reactivation occurs, Avacopan and any concomitant therapies associated with HBV reactivation must be discontinued immediately, and expert consultation sought.[7]

Serious Infections:

Serious infections, including some that were fatal, have been reported in patients receiving Avacopan. The most commonly reported serious infections in the Avacopan group in clinical trials were pneumonia and urinary tract infections.7

  • Active Infections: Use of Avacopan should be avoided in patients with an active, serious infection, including localized infections.[7]
  • Risk Factors: The risks and benefits of initiating Avacopan should be carefully considered in patients with chronic infections, those at an increased risk of infection, or those who have resided in or traveled to areas where certain infections are endemic.[7]
  • Hematological Monitoring and Thresholds: Treatment with Avacopan must be temporarily stopped if a patient develops leukopenia (white blood cell count < 2×109/L), neutropenia (absolute neutrophil count < 1×109/L), or lymphopenia (lymphocyte count < 0.2×109/L), or if an active, serious infection occurs. Avacopan treatment should not be initiated if the baseline white blood cell count is < 3.5×109/L, neutrophil count is < 1.5×109/L, or lymphocyte count is < 0.5×109/L.[9]

The severe infection rate reported in a UK real-world evidence cohort was 19.2 per 100 patient-years.23

Potential for Malignancy:

As immunomodulatory medicinal products may theoretically increase the risk for malignancies, this is a consideration. However, clinical data on Avacopan in this regard are currently limited.9

Cardiac Disorders:

Patients with GPA or MPA are inherently at risk of cardiac disorders such as myocardial infarction, cardiac failure, and cardiac vasculitis. Serious adverse events of cardiac disorder have been reported in patients treated with Avacopan. It has been suggested that treatment regimens based on cyclophosphamide followed by azathioprine may carry an increased risk for cardiac disorders compared to regimens based on rituximab when used with Avacopan.9

These warnings and precautions, especially concerning hepatotoxicity and HBV reactivation, mandate meticulous patient selection and diligent, ongoing monitoring. These are significant safety considerations that form an integral part of the clinical management strategy for patients prescribed Avacopan. The risk of serious infections, while a known complication of most immunosuppressive therapies for AAV, also remains a key area for vigilance.

[5.3. Contraindications]

Avacopan is contraindicated in patients with:

  • A history of serious hypersensitivity to Avacopan or to any of its excipients.[8]
  • Cirrhosis: Several sources indicate that Avacopan is not recommended for patients with cirrhosis, and it is listed as a contraindication in some drug information resources.[7]

Additionally, while not always listed as an absolute contraindication, the concomitant use of strong CYP3A4 inducers (e.g., rifampin, carbamazepine) with Avacopan is strongly advised against or to be avoided due to the significant reduction in Avacopan exposure and potential loss of efficacy.[2] This effectively functions as a practical contraindication in many clinical scenarios. (It should be noted that one source [10] inaccurately listed strong CYP3A4

inhibitors as a contraindication alongside cirrhosis; however, multiple other sources and prescribing information clarify that strong CYP3A4 inhibitors necessitate a dose reduction of Avacopan, not contraindication).

6. Dosage and Administration

[6.1. Recommended Dosage for AAV]

The recommended dosage of Avacopan for adult patients with severe active ANCA-associated vasculitis (GPA and MPA) is 30 mg, administered as three 10 mg capsules, taken orally twice daily (typically morning and evening).[2]

[6.2. Method of Administration]

  • Avacopan should be administered with food. This is due to the significant increase in drug absorption observed when taken with a meal.[2]
  • The capsules must be swallowed whole with water. They should not be crushed, chewed, or opened, as this may alter the drug's release and absorption characteristics.[2]
  • Patients should be advised to avoid consuming grapefruit and grapefruit juice while being treated with Avacopan, as grapefruit is a known strong inhibitor of CYP3A4 and can increase Avacopan plasma concentrations.[9]

[6.3. Missed Dose Instructions]

If a patient misses a dose of Avacopan, they should be instructed to wait until their usual scheduled time to take the next regular dose. The patient should not take an extra dose or double the next dose to make up for the missed one.[2]

[6.4. Dosage Adjustments]

A specific dosage adjustment for Avacopan is required in the context of drug interactions:

  • Concomitant use with Strong CYP3A4 Inhibitors: When Avacopan is co-administered with strong inhibitors of the CYP3A4 enzyme (e.g., itraconazole, ketoconazole, clarithromycin, ritonavir-boosted protease inhibitors), the dosage of Avacopan must be reduced to 30 mg (three 10 mg capsules) taken once daily.[2]

The specific instructions for administration—such as taking with food, swallowing capsules whole, and avoiding grapefruit—are critical for ensuring patient adherence and achieving consistent, predictable drug exposure. The significant food effect, leading to a 72% increase in AUC [15], necessitates co-administration with food for optimal bioavailability. The dose reduction when used with strong CYP3A4 inhibitors is a direct consequence of Avacopan's primary metabolic pathway via CYP3A4 and is a vital measure to prevent potential toxicity resulting from excessive drug exposure due to inhibited metabolism.

Table 4: Dosage Recommendations and Adjustments for Avacopan

Condition / SituationRecommended Avacopan DosageAdministration NotesSource(s)
Standard Adult Dose for AAV30 mg (three 10 mg capsules) BIDWith food; swallow whole; avoid grapefruit2
Co-administration with Strong CYP3A4 Inhibitor30 mg (three 10 mg capsules) ONCE DAILYWith food; swallow whole; avoid grapefruit2

7. Drug Interactions

Avacopan's pharmacokinetic profile indicates a significant potential for drug-drug interactions (DDIs), primarily due to its metabolism and effects on the cytochrome P450 (CYP) enzyme system. Avacopan is principally metabolized by CYP3A4.[9] Furthermore, Avacopan itself is an inhibitor of CYP3A4.[1] Its active M1 metabolite may also contribute to interactions by inhibiting CYP2C9 and CYP3A4, and potentially inducing CYP3A4, CYP1A2, and CYP2B6.[15] This complex interplay necessitates careful review of concomitant medications.

[7.1. Effects of Other Drugs on Avacopan]

The plasma concentrations and therapeutic effect of Avacopan can be significantly altered by drugs that modulate CYP3A4 activity.

  • Strong and Moderate CYP3A4 Inducers:
  • Examples: Rifampin, carbamazepine, phenobarbital, phenytoin, enzalutamide, mitotane, St. John's Wort.[9]
  • Effect: These agents can significantly decrease Avacopan plasma concentrations, potentially leading to a loss of efficacy. Co-administration of Avacopan with rifampin, a strong CYP3A4 inducer, resulted in an approximate 93% decrease in Avacopan AUC and a 79% decrease in Cmax​.[9]
  • Management: Concomitant use of Avacopan with strong or moderate CYP3A4 inducers should be avoided.[2]
  • Strong CYP3A4 Inhibitors:
  • Examples: Itraconazole, ketoconazole, posaconazole, voriconazole, clarithromycin, telithromycin, cobicistat, ritonavir, and other ritonavir-boosted protease inhibitors.[15]
  • Effect: These agents can significantly increase Avacopan plasma concentrations, raising the risk of dose-related adverse effects. Co-administration with itraconazole, a strong CYP3A4 inhibitor, increased Avacopan AUC by approximately 119% (a 2.2-fold increase) and Cmax​ by 87%.[15]
  • Management: When Avacopan is used concomitantly with a strong CYP3A4 inhibitor, its dosage must be reduced to 30 mg once daily.[2]
  • Grapefruit and Grapefruit Juice:
  • Effect: Grapefruit juice is a well-known strong inhibitor of intestinal CYP3A4 and can increase the bioavailability and plasma concentrations of CYP3A4 substrates like Avacopan.[28]
  • Management: Patients should be advised to avoid consuming grapefruit and grapefruit juice during treatment with Avacopan.[9]

[7.2. Effects of Avacopan on Other Drugs]

As an inhibitor of CYP3A4, Avacopan can increase the plasma concentrations of co-administered drugs that are substrates of this enzyme.

  • CYP3A4 Substrates:
  • Examples: Midazolam, simvastatin, atorvastatin, lovastatin, cyclosporine, tacrolimus, sirolimus, certain calcium channel blockers, and many others.
  • Effect: Avacopan is considered a moderate inhibitor of CYP3A4.[25] Co-administration can lead to increased plasma concentrations and AUC of CYP3A4 substrates, potentially increasing their therapeutic effects or risk of toxicity.[2] For example, co-administration of Avacopan with midazolam (a sensitive CYP3A4 substrate) increased midazolam AUC by 81%.[15]
  • Management: When Avacopan is co-administered with CYP3A4 substrates, a dose reduction of the CYP3A4 substrate should be considered. The prescribing information for the concomitant CYP3A4 substrate should be consulted.[2]
  • Specific recommendation for Simvastatin: Co-administration of Avacopan with 40 mg simvastatin increases simvastatin exposure. While taking Avacopan, the dosage of simvastatin should be limited to 10 mg daily. For patients who have previously tolerated simvastatin 80 mg daily for at least one year without evidence of muscle toxicity, a simvastatin dose of 20 mg daily may be considered with Avacopan.[7]
  • CYP2C9 Substrates:
  • Effect: The M1 metabolite of Avacopan may inhibit CYP2C9.[15]
  • Management: An interaction study with celecoxib (a CYP2C9 substrate) showed increased celecoxib exposure, but this was not expected to be clinically important, and no specific management was recommended.[15] However, caution may be warranted with other sensitive CYP2C9 substrates, particularly those with a narrow therapeutic index.
  • Proton Pump Inhibitors (PPIs):
  • Effect: Drugs affecting gastric acidity, such as omeprazole (a PPI), are not expected to have a clinically significant effect on Avacopan pharmacokinetics.[15]

The extensive drug interaction profile of Avacopan, stemming from its role as both a substrate and an inhibitor of CYP3A4, necessitates a thorough review of all concomitant medications (including prescription, over-the-counter, and herbal products) before initiating and during Avacopan therapy. The interactions with strong CYP3A4 inducers (risking therapeutic failure of Avacopan) and strong CYP3A4 inhibitors (risking Avacopan toxicity) are particularly critical and have clearly defined management strategies. Similarly, Avacopan's potential to elevate levels of other CYP3A4 substrates, such as statins, requires proactive dose adjustments or careful monitoring. The extensive list of potential interactants underscores the importance of vigilance from both prescribing physicians and pharmacists.

Table 5: Clinically Significant Drug Interactions with Avacopan and Management Strategies

Interacting Drug/ClassEffect on Avacopan or Interacting DrugClinical ConsequenceManagement RecommendationSource(s)
Strong/Moderate CYP3A4 Inducers (e.g., rifampin, carbamazepine, St. John's Wort)↓ Avacopan exposure (AUC by ~93% with rifampin)Potential loss of Avacopan efficacyAvoid concomitant use2
Strong CYP3A4 Inhibitors (e.g., itraconazole, ketoconazole, clarithromycin, ritonavir)↑ Avacopan exposure (AUC by ~119% with itraconazole)Potential increased Avacopan toxicityReduce Avacopan dosage to 30 mg once daily2
Grapefruit / Grapefruit Juice↑ Avacopan exposure (predicted)Potential increased Avacopan toxicityAvoid consumption9
CYP3A4 Substrates (general)↑ Exposure of CYP3A4 substrate (e.g., midazolam AUC ↑ by 81%)Potential increased toxicity/effects of the substrateConsider dose reduction of the CYP3A4 substrate; consult substrate's prescribing information2
Simvastatin (CYP3A4 substrate)↑ Simvastatin exposureIncreased risk of simvastatin-related adverse effects (e.g., myopathy)Limit simvastatin dosage to 10 mg daily (or 20 mg daily for certain previously tolerant patients) with Avacopan co-administration7

8. Use in Specific Populations

[8.1. Hepatic Impairment]

Avacopan is primarily metabolized in the liver by CYP3A4, making its use in patients with hepatic impairment a key consideration.

  • Mild (Child-Pugh Class A) or Moderate (Child-Pugh Class B) Hepatic Impairment: Pharmacokinetic studies have shown that in patients with mild or moderate hepatic impairment, Avacopan AUC increased by approximately 12%, while Cmax​ decreased slightly. Changes in the exposure of the M1 metabolite were also minimal. These alterations are not considered clinically important, and thus, dose adjustment of Avacopan is generally not deemed necessary in patients with mild to moderate hepatic impairment.[10]
  • Severe (Child-Pugh Class C) Hepatic Impairment: Avacopan has not been studied in patients with severe hepatic impairment.[15]
  • Recommendations for Liver Disease/Cirrhosis: Given Avacopan's known potential for hepatotoxicity (see Section 5.2), it is not recommended for patients with active, untreated, and/or uncontrolled chronic liver disease, such as chronic active hepatitis B, untreated hepatitis C, or uncontrolled autoimmune hepatitis. It is also not recommended for patients with cirrhosis.[7] Some sources explicitly list cirrhosis as a contraindication.[10]

While mild to moderate hepatic impairment does not appear to significantly alter Avacopan's pharmacokinetics, the absence of data in severe impairment, combined with the established warnings regarding hepatotoxicity and specific non-recommendations or contraindications for active liver disease and cirrhosis, dictates a very cautious approach. Avacopan should generally be avoided or used with extreme caution and intensive monitoring in patients with any significant underlying liver dysfunction.

[8.2. Renal Impairment]

The influence of renal impairment on Avacopan's pharmacokinetics has been evaluated.

  • Studies indicate that for patients with an estimated glomerular filtration rate (eGFR) ranging from 14 to 170 mL/minute per 1.73 m², there is no clinically important effect on Avacopan pharmacokinetics.[15] This suggests that standard doses can likely be used in patients with mild, moderate, or severe renal impairment not requiring dialysis.
  • It is important to note that patients with an eGFR below 15 mL/min/1.73 m² were excluded from the pivotal ADVOCATE trial.[23] Consequently, prospective, controlled efficacy and safety data for this specific subgroup with very severe renal dysfunction are lacking from the primary evidence base supporting Avacopan's approval.
  • However, emerging real-world evidence (RWE) includes patients with eGFR <15 mL/min/1.73 m², and some reports suggest clinical benefit in this population, including those dependent on dialysis.[11] This use is currently outside the scope of the ADVOCATE trial's population but reflects an area of active clinical exploration.

Based on pharmacokinetic data, dose adjustments for renal impairment (down to eGFR 14 mL/min/1.73 m²) do not appear necessary. The use in patients with eGFR <15 mL/min/1.73 m² or those on dialysis should be guided by evolving RWE and expert opinion, acknowledging the limitations of controlled trial data in this specific population.

[8.3. Pediatric Use]

The safety and efficacy of Avacopan in pediatric patients (under 18 years of age) were not established in the initial clinical trials leading to its approval for adults.[26]

  • The European Medicines Agency (EMA) has noted a Paediatric Investigation Plan (PIP) decision for Avacopan, which includes considerations for age-appropriate oral solid and liquid dosage forms.[3]
  • Currently, an ongoing Phase 3, open-label, single-arm clinical trial (NCT06321601 / Amgen study 20230070) is actively evaluating the efficacy, pharmacokinetics, and safety of Avacopan in children aged 6 years to <18 years with active AAV. In this study, Avacopan is administered in combination with either a rituximab-containing or a cyclophosphamide-containing regimen. The trial aims to enroll approximately 20 participants, with an estimated primary completion date of July 2030.[29]
  • A case report has described rapid Avacopan-induced liver injury in a 10-year-old child, highlighting that safety concerns, particularly hepatotoxicity, will also be relevant in the pediatric population.[10]

Avacopan is not yet approved for pediatric use. The ongoing Phase 3 trial is a critical step towards potentially extending its indication to children with AAV. The results of this study will be essential for establishing appropriate dosing, efficacy, and safety in this younger age group.

[8.4. Geriatric Use]

AAV predominantly affects older adults, making the use of Avacopan in the geriatric population highly relevant.

  • Clinical studies of Avacopan, including the ADVOCATE trial (where the mean age was approximately 60 years), have included a significant number of elderly patients.[18]
  • Available data to date have not demonstrated specific geriatric problems that would limit the usefulness of Avacopan in older adults.[26] Standard adult dosing is generally applicable, with the usual clinical considerations for age-related changes in organ function, potential for polypharmacy, and management of comorbidities.

[8.5. Pregnancy and Lactation]

Data on the use of Avacopan during pregnancy and lactation are limited.

  • Pregnancy: It is currently unknown whether Avacopan (Tavneos) may cause harm to an unborn baby if used during pregnancy.[2] Animal reproduction studies, if available, would be detailed in the full prescribing information. Avacopan has an Australian Pregnancy Category D classification [1], which indicates that there is evidence of an increased risk of human fetal damage, but the benefits from use in pregnant women may be acceptable despite the risk (e.g., if the drug is needed in a life-threatening situation or for a serious disease for which safer drugs cannot be used or are ineffective).
  • Lactation: It is not known whether Avacopan or its metabolites pass into human breast milk.[2] The potential effects on a breastfed infant are therefore unknown.

Given the insufficient data, a careful risk-benefit assessment is required if considering Avacopan use in pregnant or breastfeeding women. Alternative therapies with more established safety profiles in these situations might be preferred. The Australian Category D designation suggests caution and implies that use during pregnancy should only be considered if the potential benefits clearly justify the potential risks to the fetus.

9. Regulatory Status and Approvals

Avacopan (Tavneos) has received regulatory approvals in several major jurisdictions worldwide for the treatment of ANCA-associated vasculitis, reflecting a global recognition of its clinical value.

  • Orphan Drug Designations: Avacopan received orphan drug designations from both the FDA (for ANCA-associated vasculitis and C3G) and the EMA (for GPA, MPA, and C3 glomerulopathy).[3] These designations are granted to drugs intended for rare diseases and provide incentives to support their development.

The rapid succession of approvals across these key regulatory agencies between late 2021 and mid-2022 underscores the strength of the clinical data package for Avacopan and the recognized unmet medical need in AAV. The orphan drug status likely facilitated and accelerated its development pathway. While the core indication for GPA and MPA is consistent, slight variations in the specific wording of the indications exist between regions (e.g., the FDA's explicit mention of use "including glucocorticoids" versus the EMA's phrasing "glucocorticoids as clinically indicated").

Table 6: Summary of Global Regulatory Approvals for Avacopan (Tavneos)

Regulatory AgencyCountry/RegionApproval Date(s)Approved Indication for AAV (Adults)Trade Name / Marketing Authorization Holder(s)Source(s)
FDAUSAOct 7/8, 2021Adjunctive treatment of severe active GPA and MPA in combination with standard therapy including glucocorticoids.TAVNEOS / ChemoCentryx (later Amgen)User Query, 1
EMAEuropean UnionJan 19, 2022Treatment of severe, active GPA or MPA in combination with a rituximab or cyclophosphamide regimen (glucocorticoids as clinically indicated).TAVNEOS / Vifor Fresenius Medical Care Renal Pharma (VFMCRP) / CSL ViforUser Query, 1
MHRAUnited KingdomMay 2022Treatment of severe, active GPA or MPA in combination with a rituximab or cyclophosphamide regimen.TAVNEOS / VFMCRP / CSL Vifor32
PMDAJapanSep 27, 2021Treatment of MPA and GPA.TAVNEOS / Kissei Pharmaceutical Co., Ltd.1
Health CanadaCanadaApr 20, 2022Adjunctive treatment of severe active GPA and MPA in combination with standard therapy including glucocorticoids.TAVNEOSUser Query, 3

10. Cost-Effectiveness

The cost-effectiveness of Avacopan has been evaluated by various health technology assessment (HTA) bodies, with conclusions varying by jurisdiction, largely influenced by local willingness-to-pay (WTP) thresholds, healthcare system costs, and, critically, the negotiated price of the drug.

  • Canada (CADTH): A pharmacoeconomic review by the Canadian Agency for Drugs and Technologies in Health (CADTH) concluded that Avacopan plus standard of care (SOC) compared to SOC alone resulted in an incremental cost-effectiveness ratio (ICER) of $365,453 per quality-adjusted life-year (QALY) gained, based on the CADTH base case analysis.[27] At a commonly cited WTP threshold of $50,000 per QALY, Avacopan was not considered cost-effective. CADTH estimated that a price reduction of 72.5% would be required for Avacopan to meet this threshold. The analysis highlighted uncertainties related to the use of non-study glucocorticoids in the trial, relapse rates, and the lack of long-term clinical data. The annual list price of Avacopan in Canada was cited as $75,051 CAD.[27]
  • Spain: A cost-utility analysis conducted from the Spanish national healthcare system perspective found that Avacopan in combination with rituximab or cyclophosphamide, compared to a glucocorticoid-based regimen, yielded an ICER of €45,638 per QALY gained.[21] This was considered cost-effective within a WTP threshold of €60,000 per QALY. The model suggested that Avacopan was associated with a lower incidence of end-stage renal disease (ESRD), fewer relapses, and reduced hospitalization-related adverse events. The incremental cost of Avacopan was €16,009, with an incremental QALY gain of 0.35 (6.52 vs. 6.17 QALYs).[21]
  • United Kingdom (NICE): The National Institute for Health and Care Excellence (NICE) has recommended Avacopan for routine use on the NHS for treating severe, active GPA or MPA in adults, in combination with rituximab or cyclophosphamide.[21] The list price was £5,547.95 per pack of 180 x 10 mg capsules, equating to an approximate annual cost of £67,311.[32] NICE's positive recommendation is contingent upon a confidential commercial arrangement (a simple discount patient access scheme) that makes Avacopan available to the NHS at a reduced price.[32] NICE acknowledged that Avacopan plus rituximab was more effective than standard treatment at preventing disease worsening.[32]
  • USA: At the time of its launch in the US, the expected annual wholesale acquisition cost (WAC) for Tavneos was reported to be between $150,000 and $200,000 USD.[36]

Key drivers influencing the cost-effectiveness of Avacopan include its potential to reduce the progression to ESRD (a major long-term cost for healthcare systems), decrease the frequency of disease relapses and associated hospitalizations, and lower the costs related to managing glucocorticoid-induced adverse events.[21] While Avacopan leads to higher drug acquisition costs [21], the QALYs gained are attributed to improved sustained remission rates and reduced glucocorticoid toxicity.

The variability in cost-effectiveness assessments across different countries highlights that while Avacopan offers clear clinical benefits, its high acquisition cost means that its economic value is heavily dependent on the price negotiated within each healthcare system and the specific WTP thresholds employed. The long-term economic benefits, such as averted costs from reduced ESRD incidence and fewer complications from chronic glucocorticoid use, are crucial components of its overall value proposition that aim to offset the upfront drug expenditure.

11. Ongoing Research and Future Directions

The clinical development and investigation of Avacopan continue, with research focusing on long-term outcomes in AAV, its use in pediatric populations, and its potential efficacy in other complement-mediated diseases.

  • 11.1. Long-term Efficacy and Safety Studies: Data on the use of Avacopan beyond the 52-week duration of the ADVOCATE trial are actively emerging from Early Access Programs (EAPs) and real-world evidence (RWE) studies.23 These initial reports suggest a manageable safety profile and the potential for sustained disease control with treatment extending beyond one year.24 For instance, an analysis from an EAP with a median treatment duration of 16 months in 59 patients indicated continued disease control with acceptable safety.24 Ongoing analyses from RWE cohorts, such as the UK study, are expected to provide further insights into longer-term outcomes and the effects observed after Avacopan discontinuation.23
  • 11.2. Potential New Indications: The C5a-C5aR pathway is implicated in the pathophysiology of various inflammatory and autoimmune conditions beyond AAV. Consequently, Avacopan is being, or has been, investigated for several potential new indications:
  • C3 Glomerulopathy (C3G): Avacopan has been studied in a Phase 2 clinical trial (NCT03301467) for C3G, a rare and severe kidney disease. It has received orphan drug designation for C3G from both the FDA and EMA.[13]
  • Hidradenitis Suppurativa (HS): This chronic inflammatory skin disorder has also been an area of investigation for Avacopan (e.g., trial NCT03852472), and it received orphan drug designation for HS in the past, though current development status may vary.[29]
  • Lupus Nephritis (LN): Avacopan has been explored for the treatment of lupus nephritis.[31]
  • IgA Nephropathy (IgAN): Clinical trials are investigating Avacopan in patients with IgAN (e.g., NCT05988008, NCT05988034).[29]
  • Cardiac Safety (QT Interval): Studies have also been conducted to assess Avacopan's effect on cardiac repolarization, specifically the QT interval.[29]
  • 11.3. Pediatric Clinical Trials: As detailed in Section 8.3, a Phase 3 clinical trial (NCT06321601 / Amgen study 20230070) is currently underway to evaluate the efficacy, pharmacokinetics, and safety of Avacopan in children aged 6 to <18 years with active AAV.29 This trial is crucial for potentially extending Avacopan's indication to the pediatric AAV population, addressing an important unmet medical need.

The ongoing research landscape for Avacopan is dynamic. Beyond optimizing its application in AAV through long-term follow-up and pediatric studies, there is considerable interest in harnessing its specific C5aR antagonistic mechanism for other diseases where the complement system plays a pathogenic role. Success in these additional indications, particularly in renal and dermatological conditions, could significantly broaden Avacopan's therapeutic utility and impact.

12. Conclusion and Clinical Positioning

[12.1. Summary of Avacopan's Role in AAV Management]

Avacopan (Tavneos) is a novel, first-in-class, orally administered selective C5a receptor antagonist that represents a significant advancement in the therapeutic armamentarium for ANCA-associated vasculitis (AAV).[1] Its development and approval address a long-standing need for more targeted therapies that can effectively control disease activity while minimizing the substantial toxicities associated with conventional treatments, particularly high-dose and prolonged glucocorticoid use. As demonstrated in the pivotal ADVOCATE trial, Avacopan, when used as adjunctive therapy, offers effective disease control, especially in sustaining remission over the longer term.[4]

[12.2. Benefits]

The introduction of Avacopan into AAV management offers several key benefits:

  • Glucocorticoid-Sparing: The most prominent benefit is its ability to significantly reduce the cumulative exposure to glucocorticoids and, consequently, their associated toxicities. This is a major clinical advantage, as glucocorticoid-related adverse effects are a primary source of morbidity in the long-term management of AAV.[11]
  • Sustained Remission: Avacopan demonstrated superiority over a standard prednisone taper regimen in maintaining disease remission at 52 weeks in the ADVOCATE trial.[4]
  • Renal Benefits: Clinical trial data suggest potential for improved renal outcomes, including greater improvement in eGFR and a faster reduction in albuminuria, which are particularly relevant given the frequent and severe kidney involvement in AAV.[11]
  • Improved Health-Related Quality of Life (HRQoL): Treatment with Avacopan has been associated with better HRQoL outcomes compared to standard prednisone-based regimens.[11]
  • Oral Administration: As an oral medication, Avacopan offers convenience for patients compared to intravenously administered therapies.[1]

[12.3. Key Considerations for Clinical Use]

While Avacopan offers significant advantages, its optimal and safe use requires attention to several key considerations:

  • Adjunctive Therapy: It is approved for use in combination with standard AAV therapies, typically rituximab or cyclophosphamide, and does not entirely replace the need for glucocorticoids in all initial treatment phases as per some regional indications.[1]
  • Safety Monitoring: Due to the identified risk of hepatotoxicity, regular monitoring of liver function tests is mandatory before and during treatment. Vigilance for signs of HBV reactivation (requiring pre-screening) and serious infections is also crucial.[7]
  • Drug Interactions: Avacopan has a high potential for clinically significant drug-drug interactions due to its metabolism by and inhibition of CYP3A4. A thorough review of concomitant medications and appropriate dose adjustments are essential.[9]
  • Patient Selection: Avacopan may be particularly valuable for patients at high risk of glucocorticoid-related toxicity (e.g., those with diabetes, osteoporosis, psychiatric conditions, or high infection risk), individuals with significant renal involvement (especially those with rapidly declining kidney function), or potentially those with a history of relapsing disease.[11]

[12.4. Positioning Relative to Standard of Care]

Avacopan is increasingly recognized in international treatment guidelines as an important option for the remission induction phase of severe, active GPA and MPA, specifically to facilitate a substantial reduction in the glucocorticoid burden.[11] It offers an alternative to treatment strategies reliant on prolonged high-dose steroid regimens, aiming to shift the risk-benefit balance by minimizing steroid-induced harm while maintaining or improving therapeutic efficacy.

The core problem with traditional AAV treatment has long been the heavy reliance on glucocorticoids and their extensive, often debilitating, side effect profile. Avacopan's primary achievement in the ADVOCATE trial was demonstrating sustained disease remission with significantly less glucocorticoid use and a corresponding reduction in glucocorticoid-related toxicity. This directly addresses this central challenge in AAV care. Consequently, treatment guidelines are incorporating Avacopan as a glucocorticoid-sparing agent.

Therefore, Avacopan is not merely an additional drug but a strategic therapeutic tool that has the potential to fundamentally alter the approach to AAV treatment, steering it towards more targeted and less broadly toxic regimens. It allows for a more favorable long-term outlook for many patients. However, its own safety profile, particularly the need for liver function monitoring, HBV screening, vigilance for infections, and careful management of drug interactions, means that its use requires informed clinical judgment and diligent patient management. It represents a trade-off, aiming to replace the well-known risks of glucocorticoids with a different, and hopefully more manageable, set of potential risks associated with a targeted therapy. The optimal duration of Avacopan therapy, its potential role in achieving complete steroid avoidance in selected patients from treatment initiation, and its utility in long-term maintenance therapy are areas where clinical experience and ongoing research, including real-world evidence generation, will continue to refine its position in the AAV treatment landscape. The long-term impact of Avacopan on preventing cumulative organ damage and improving overall morbidity and mortality in AAV will become clearer with continued follow-up studies and broader clinical use.

Works cited

  1. Avacopan - Wikipedia, accessed June 13, 2025, https://en.wikipedia.org/wiki/Avacopan
  2. Tavneos: Uses, Dosage, Side Effects & Warnings - Drugs.com, accessed June 13, 2025, https://www.drugs.com/tavneos.html
  3. Avacopan | C33H35F4N3O2 | CID 49841217 - PubChem, accessed June 13, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Avacopan
  4. Avacopan (TAVNEOS) in ANCA-positive Granulomatosis with Polyangiitis and Microscopic Polyangiitis National Drug Monograph August - VA.gov, accessed June 13, 2025, https://www.va.gov/formularyadvisor/DOC_PDF/MON_Avacopan_TAVNEOS_in_GPA_MPA_Monograph_Aug_2022.pdf
  5. Avacopan: Uses, Dosage, Side Effects, Warnings - Drugs.com, accessed June 13, 2025, https://www.drugs.com/avacopan.html
  6. Avacopan (CCX168) | CAS 1346623-17-3 | AbMole BioScience, accessed June 13, 2025, https://www.abmole.com/products/avacopan.html
  7. Dosing and Administration | TAVNEOS® (avacopan), accessed June 13, 2025, https://www.tavneospro.com/about-tavneos/dosing
  8. Safety Profile | TAVNEOS® (avacopan), accessed June 13, 2025, https://www.tavneospro.com/results/safety
  9. Tavneos, INN-avacopan - EMA, accessed June 13, 2025, https://www.ema.europa.eu/documents/product-information/tavneos-epar-product-information_de.pdf
  10. Avacopan - LiverTox - NCBI Bookshelf, accessed June 13, 2025, https://www.ncbi.nlm.nih.gov/books/NBK609016/
  11. Evaluating avacopan in the treatment of ANCA-Associated vasculitis ..., accessed June 13, 2025, https://www.dovepress.com/evaluating-avacopan-in-the-treatment-of-anca-associated-vasculitis-des-peer-reviewed-fulltext-article-DDDT
  12. Avacopan | CAS#:1346623-17-3 | Chemsrc, accessed June 13, 2025, https://www.chemsrc.com/en/cas/1346623-17-3_1276477.html
  13. avacopan | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed June 13, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=9450
  14. アバコパン | New Drug Approvals, accessed June 13, 2025, https://newdrugapprovals.org/tag/%E3%82%A2%E3%83%90%E3%82%B3%E3%83%91%E3%83%B3/
  15. Avacopan Monograph for Professionals - Drugs.com, accessed June 13, 2025, https://www.drugs.com/monograph/avacopan.html
  16. In Progress: Avacopan Pathway, Pharmacokinetics/Pharmacodynamics - PharmGKB, accessed June 13, 2025, https://www.pharmgkb.org/pathway/PA166344361/components
  17. (PDF) Evaluating Avacopan in the Treatment of ANCA-Associated ..., accessed June 13, 2025, https://www.researchgate.net/publication/387720063_Evaluating_Avacopan_in_the_Treatment_of_ANCA-Associated_Vasculitis_Design_Development_and_Positioning_of_Therapy
  18. Efficacy and safety of avacopan in patients with ANCA-associated ..., accessed June 13, 2025, https://ard.bmj.com/content/83/2/223
  19. Avacopan for the Treatment of ANCA-Associated Vasculitis - PubMed, accessed June 13, 2025, https://pubmed.ncbi.nlm.nih.gov/33596356/
  20. Drug Trials Snapshot: TAVNEOS - FDA, accessed June 13, 2025, https://www.fda.gov/drugs/drug-approvals-and-databases/drug-trials-snapshot-tavneos
  21. Full article: A projected cost-utility analysis of avacopan for the ..., accessed June 13, 2025, https://www.tandfonline.com/doi/full/10.1080/14737167.2023.2297790
  22. POS0246 DATA FROM THE ADVOCATE TRIAL ON 28 PATIENTS WITH ANCA-ASSOCIATED VASCULITIS WHO RECEIVED AVACOPAN WITHOUT CONCOMITANT GLUCOCORTICOID USE IN THE FIRST 29 DAYS | Annals of the Rheumatic Diseases, accessed June 13, 2025, https://ard.bmj.com/content/83/Suppl_1/430
  23. OA30 Real-world use and outcomes of avacopan for ANCA ..., accessed June 13, 2025, https://academic.oup.com/rheumatology/article/64/Supplement_3/keaf142.030/8115210
  24. #2256 Safety and effectiveness of avacopan beyond 52 weeks ..., accessed June 13, 2025, https://academic.oup.com/ndt/article/39/Supplement_1/gfae069-1249-2256/7678006
  25. Tavneos (Avacopan Capsules): Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed June 13, 2025, https://www.rxlist.com/tavneos-drug.htm
  26. Avacopan (oral route) - Mayo Clinic, accessed June 13, 2025, https://www.mayoclinic.org/drugs-supplements/avacopan-oral-route/description/drg-20524765
  27. Pharmacoeconomic Review - Avacopan (Tavneos) - NCBI Bookshelf, accessed June 13, 2025, https://www.ncbi.nlm.nih.gov/books/NBK598225/
  28. Avacopan | Interactions | BNF - NICE, accessed June 13, 2025, https://bnf.nice.org.uk/interactions/avacopan/
  29. Avacopan – Application in Therapy and Current Clinical Research, accessed June 13, 2025, https://clinicaltrials.eu/inn/avacopan/
  30. Study to Evaluate Avacopan in Combination with a ... - AMGEN, accessed June 13, 2025, https://www.amgentrials.com/study/?id=20230070
  31. ChemoCentryx Announces Approval in Japan of TAVNEOS ..., accessed June 13, 2025, https://www.biospace.com/chemocentryx-announces-approval-in-japan-of-tavneos-avacopan-for-the-treatment-of-anca-associated-vasculitisapproved-for-use-in-japan-in-patients-with-microscopic-polyangiitis-and-granulomatosis-with-polyangiitis
  32. CSL Vifor plans UK launch after NICE backs rare disease drug ..., accessed June 13, 2025, https://pharmaphorum.com/news/csl-vifor-plans-uk-launch-after-nice-backs-rare-disease-drug-tavneos
  33. Avacopan for treating severe active granulomatosis with polyangiitis or microscopic polyangiitis [ID1581] - NICE, accessed June 13, 2025, https://www.nice.org.uk/guidance/ta825/documents/1
  34. www.nice.org.uk, accessed June 13, 2025, https://www.nice.org.uk/guidance/ta825/documents/final-appraisal-determination-document#:~:text=2.3%20Avacopan%20costs%20%C2%A35%2C547.95,the%20NHS%20with%20a%20discount.
  35. 2 Information about avacopan | Avacopan for treating severe active granulomatosis with polyangiitis or microscopic polyangiitis | Guidance | NICE, accessed June 13, 2025, https://www.nice.org.uk/guidance/ta825/chapter/2-Information-about-avacopan
  36. ChemoCentryx nabs FDA approval of Tavneos for ANCA-associated vasculitis, accessed June 13, 2025, https://firstwordpharma.com/story/5415603
  37. Tavneos (avacopan) FDA Approval History - Drugs.com, accessed June 13, 2025, https://www.drugs.com/history/tavneos.html

Published at: June 13, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.