MedPath

ibuzatrelvir Advanced Drug Monograph

Published:Oct 31, 2025

Generic Name

ibuzatrelvir

Ibuzatrelvir (PF-07817883): A Comprehensive Profile of a Second-Generation Oral Antiviral for the Treatment of COVID-19

Executive Summary

Ibuzatrelvir, also known by its development code PF-07817883, is an investigational, orally bioavailable antiviral agent developed by Pfizer as a second-generation therapeutic for the treatment of COVID-19.[1] It is classified as a small molecule inhibitor of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro), an enzyme essential for viral replication. The primary strategic rationale behind its development is to improve upon the clinical limitations of its predecessor, nirmatrelvir, the active antiviral component of the combination therapy Paxlovid.[1]

The mechanism of action of ibuzatrelvir involves a reversible covalent interaction between its nitrile "warhead" and the active site cysteine residue (Cys145) of the viral 3C-like (3CL) protease.[1] This interaction effectively blocks the enzyme's ability to cleave viral polyproteins, thereby halting the viral replication cycle. A key design feature of ibuzatrelvir is its improved metabolic stability compared to nirmatrelvir. This enhancement obviates the need for co-administration with a pharmacokinetic booster like ritonavir, which is a potent inhibitor of the cytochrome P450 3A (CYP3A) enzyme system.[3] By eliminating ritonavir from the regimen, ibuzatrelvir is designed to have a significantly lower potential for the drug-drug interactions (DDIs) that currently limit the use of Paxlovid in patients with complex comorbidities and concomitant medications.

The clinical development program for ibuzatrelvir has progressed rapidly, with successful completion of Phase 1 and Phase 2b trials. These studies have established a favorable safety profile, which has been shown to be comparable to placebo.[6] The Phase 2b dose-ranging study (NCT05799495) demonstrated robust, dose-dependent antiviral activity, with all tested doses (100 mg, 300 mg, and 600 mg) achieving statistically significant reductions in SARS-CoV-2 viral load compared to placebo in non-hospitalized adults with COVID-19.[6] Notably, the tolerability profile was excellent, with no reports of dysgeusia (taste disturbance), an unpleasant side effect commonly associated with Paxlovid, often referred to as "Paxlovid mouth".[1]

Currently, ibuzatrelvir is advancing through a strategically designed, pivotal Phase 3 clinical trial program. One large-scale study (NCT06679140) is evaluating its efficacy in preventing severe outcomes in the general high-risk outpatient population.[9] A second, highly targeted study (NCT07013474) is investigating its use in severely immunocompromised patients, a vulnerable population for whom the DDI profile of Paxlovid is a major clinical concern.[10] The U.S. Food and Drug Administration (FDA) has granted ibuzatrelvir Fast Track designation, underscoring its potential to address an unmet medical need.[1] Pending positive outcomes from the ongoing Phase 3 trials, ibuzatrelvir is poised to become a critical new therapeutic option for COVID-19, offering a safer and more broadly applicable treatment, particularly for patients who are currently ineligible for or have contraindications to Paxlovid.

Introduction and Chemical Profile

Introduction

Ibuzatrelvir is an investigational small molecule antiviral drug being developed by the global pharmaceutical company Pfizer for the oral treatment of coronavirus disease 2019 (COVID-19).[1] Identified by the development code PF-07817883, it represents a second-generation therapeutic in the class of SARS-CoV-2 main protease (Mpro) inhibitors.[2] Its development was undertaken as a strategic initiative to overcome the specific clinical limitations of the first-generation Mpro inhibitor, nirmatrelvir, which is the active antiviral component of the widely used combination therapy Paxlovid.[1] By engineering a molecule with enhanced metabolic stability, Pfizer aims to provide a standalone oral antiviral that does not require pharmacokinetic boosting, thereby expanding the eligible patient population and simplifying clinical management of COVID-19.

Chemical and Physical Data

The molecular structure and identity of ibuzatrelvir have been well-defined and are consistent across multiple public and commercial databases. This high degree of consistency across independent sources provides a strong foundation of confidence in the compound's chemical identity, ensuring that all subsequent pharmacological and clinical data are correctly attributed to this specific molecular entity.[1] The fundamental chemical and physical properties of ibuzatrelvir are summarized below and consolidated in Table 1.

  • Development Code: PF-07817883 [1]
  • International Union of Pure and Applied Chemistry (IUPAC) Name: Methyl N-ethyl]carbamoyl]-4-(trifluoromethyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2-yl]carbamate [1]
  • Chemical Formula: $C_{21}H_{30}F_{3}N_{5}O_{5}$ [1]
  • Molar Mass / Molecular Weight: 489.496 g·mol⁻¹ [1]
  • Chemical Abstracts Service (CAS) Number: 2755812-39-4 [1]

Table 1: Chemical and Pharmacological Identifiers of Ibuzatrelvir

IdentifierValueSource(s)
Generic NameIbuzatrelvir[14]
Development CodePF-078178831
IUPAC NameMethyl N-ethyl]carbamoyl]-4-(trifluoromethyl)pyrrolidin-1-yl]-3,3-dimethyl-1-oxobutan-2-yl]carbamate1
Chemical Formula$C_{21}H_{30}F_{3}N_{5}O_{5}$[1, 14]
Molar Mass489.496 g·mol⁻¹[1, 14]
CAS Number2755812-39-4[1, 14]
DrugBank Accession No.DB19451[14]
PubChem CID1633620001
UNIIKZ2X7QH2VT[1, 14]
InChI KeyWGNWEPPRWQKSKI-AIEDFZFUSA-N[1, 14]
SMILESCC(C)(C)C@@HNC(=O)OC

Mechanism of Action and Pharmacological Rationale

Primary Target: SARS-CoV-2 Main Protease (Mpro/3CLpro)

The primary antiviral activity of ibuzatrelvir is derived from its highly specific inhibition of the SARS-CoV-2 main protease (Mpro), an enzyme also referred to as the 3C-like protease (3CLpro). This enzyme plays an indispensable role in the viral life cycle. The SARS-CoV-2 genome is translated by the host cell's machinery into large polyproteins (pp1a and pp1ab), which are non-functional precursors. The Mpro enzyme is responsible for cleaving these polyproteins at multiple specific sites to release a cascade of functional non-structural proteins (NSPs) that are essential for forming the viral replication and transcription complex. By targeting and inhibiting Mpro, ibuzatrelvir effectively prevents the processing of these polyproteins, thereby blocking the formation of the viral replication machinery and halting the production of new virions. The high degree of conservation of the Mpro enzyme across coronaviruses and its lack of a close human homologue make it an ideal and highly validated target for antiviral drug development, minimizing the risk of off-target effects in the host.

Molecular Interaction Details

The inhibitory mechanism of ibuzatrelvir is characterized as a reversible covalent interaction with its enzymatic target. The molecule is designed with a key functional group, a nitrile "warhead" ($C \equiv N$), which acts as an electrophile. This nitrile group specifically targets the nucleophilic thiol group ($-SH$) of the cysteine residue at position 145 (Cys145) within the active site of the Mpro enzyme. The interaction results in the formation of a thioimidate adduct, a covalent bond that physically blocks the substrate-binding pocket and renders the enzyme catalytically inactive. This prevents the protease from binding to and cleaving the viral polyproteins. The "reversible" nature of this covalent bond is a critical pharmacological feature. It implies that the bond can eventually break, allowing the drug to dissociate from the enzyme. This characteristic is considered advantageous as it can reduce the potential for off-target covalent modifications of other host proteins and may lower the risk of idiosyncratic toxicity or immunogenicity that can be associated with irreversible inhibitors.

Structural Basis of Binding

High-resolution co-crystal structures of ibuzatrelvir bound to the SARS-CoV-2 Mpro have provided a detailed molecular understanding of its potent inhibitory activity. These structural studies reveal that the drug molecule fits snugly into the enzyme's active site, mimicking the natural substrate. A particularly important structural feature of ibuzatrelvir is its trifluoromethyl group ($-CF_3$). This group is precisely positioned to fit into the S2 substrate-binding subsite of the main protease. The S2 pocket of the Mpro is a deep, hydrophobic pocket that typically accommodates a leucine residue in the natural substrate. The trifluoromethyl group's size and properties allow it to establish favorable interactions within this pocket, contributing significantly to the high binding affinity and specificity of the inhibitor. This structural insight not only explains the drug's potency but also guided its rational design as an improvement over nirmatrelvir.

Pan-Coronavirus Activity

A significant pharmacological attribute of ibuzatrelvir is its demonstrated broad-spectrum activity against multiple human coronaviruses, a highly desirable feature for a therapeutic intended to combat a rapidly evolving virus and for future pandemic preparedness. The Mpro enzyme is highly conserved across the Coronaviridae family, making it a viable target for pan-coronavirus inhibitors.

  • In Vitro Potency: Preclinical studies have quantified ibuzatrelvir's potent inhibitory activity against the Mpro of several pathogenic coronaviruses. It exhibits strong inhibition of SARS-CoV-2 Mpro, with reported half-maximal inhibitory concentration ($IC_{50}$) values of 19 nM and an inhibition constant ($K_i$) of 5 nM. Its activity against the original SARS-CoV-1 Mpro is nearly identical, with a reported $IC_{50}$ of 18 nM.
  • Activity Against MERS-CoV: Ibuzatrelvir also inhibits the Mpro of the Middle East respiratory syndrome-related coronavirus (MERS-CoV), which causes a less widespread but highly lethal disease (approximately 36% mortality). The reported potency against MERS-CoV Mpro varies, with one study citing an $IC_{50}$ of 930 nM and another reporting an $IC_{50}$ of 65 nM. This discrepancy has been attributed to the use of different fluorescent resonance energy transfer (FRET) substrates in the respective assays.
  • Structural Rationale for Potency Differences: Despite the lower potency against MERS-CoV, structural studies show that ibuzatrelvir binds to the MERS-CoV Mpro active site in a very similar manner to how it binds to the SARS-CoV-2 Mpro. The observed difference in potency is rationalized by subtle variations in the substrate preferences of the two enzymes. These minor differences result in a weaker hydrogen-bond network forming between the drug and the MERS-CoV Mpro, leading to a lower overall binding affinity and faster dissociation compared to the SARS-CoV-2 complex.

The demonstrated activity of ibuzatrelvir against three distinct and highly pathogenic human coronaviruses—SARS-CoV-2, SARS-CoV-1, and MERS-CoV—positions it as more than just a COVID-19 therapeutic. It represents a potential broad-spectrum antiviral agent with utility against future coronavirus threats. The emergence of novel coronaviruses is a persistent public health risk. Having a safe, orally available antiviral with proven activity against the conserved Mpro target could be a cornerstone of future pandemic preparedness strategies, allowing for rapid deployment against a newly emerged coronavirus before specific vaccines or monoclonal antibodies can be developed. This elevates the strategic value of ibuzatrelvir beyond the immediate context of the current COVID-19 pandemic.

Strategic Development: An Enhanced Alternative to Paxlovid

Addressing the Limitations of Paxlovid

The development of ibuzatrelvir by Pfizer is a clear and deliberate strategic effort to address the well-documented clinical limitations of its first-generation Mpro inhibitor therapy, Paxlovid. Paxlovid consists of two components: the active antiviral agent, nirmatrelvir, and a pharmacokinetic boosting agent, ritonavir.

The primary challenge with nirmatrelvir is its low metabolic stability; it is rapidly broken down in the body, primarily by the cytochrome P450 3A (CYP3A) enzyme system, leading to insufficient plasma concentrations to exert a therapeutic effect when administered alone. To overcome this, Paxlovid includes ritonavir, a potent inhibitor of CYP3A enzymes. Ritonavir effectively "boosts" nirmatrelvir's exposure by slowing its metabolism, allowing it to reach and maintain therapeutic levels. However, this solution introduces a significant clinical problem: ritonavir's powerful and broad inhibition of CYP3A leads to a high potential for clinically significant drug-drug interactions (DDIs). Many patients at high risk for severe COVID-19 are older and have comorbidities for which they take multiple medications (polypharmacy). A substantial number of these common medications—including certain statins, anticoagulants, antiplatelet agents, and immunosuppressants—are also metabolized by CYP3A. Co-administration with Paxlovid can dangerously elevate the levels of these drugs, creating a risk of serious toxicity. Consequently, a large segment of the patient population that could most benefit from an oral antiviral is precluded from using Paxlovid due to these DDI concerns.

Ibuzatrelvir's Key Advantages

Ibuzatrelvir was rationally designed through targeted molecular modifications to the nirmatrelvir scaffold to directly overcome these limitations. Its development represents a classic pharmaceutical life cycle management strategy, where a company proactively creates a "bio-better" or second-generation version of its own successful product to address known weaknesses, expand the market, and extend the franchise's longevity.

  • Improved Metabolic Stability: The central innovation of ibuzatrelvir is its intrinsically enhanced metabolic stability. Pfizer's medicinal chemists achieved this by making key structural changes, such as replacing a metabolically vulnerable geminal dimethyl group on nirmatrelvir's fused bicyclic ring system with a more stable proline moiety that has a pendant trifluoromethyl group. This modification makes the molecule less susceptible to oxidation by CYP3A enzymes. As a result, ibuzatrelvir can achieve and maintain therapeutic systemic concentrations without the need for co-administration with ritonavir.
  • Reduced DDI Potential: By eliminating the ritonavir booster, ibuzatrelvir inherently possesses a significantly lower potential for DDIs. This is arguably its most important clinical advantage. It opens the door for treatment to a much broader patient population, including those with complex medical histories who are on multiple medications, such as organ transplant recipients on immunosuppressants or cancer patients undergoing chemotherapy—groups that are at very high risk for severe COVID-19 but are often poor candidates for Paxlovid.
  • Favorable Side-Effect Profile: A consistent finding from the clinical development program is an improvement in tolerability. Specifically, participants in the Phase 2b trial of ibuzatrelvir did not report experiencing dysgeusia, the unpleasant metallic or bitter taste commonly referred to as "Paxlovid mouth". While not a medically serious side effect, dysgeusia is a frequent complaint from patients taking Paxlovid and can negatively impact treatment adherence and the overall patient experience. The absence of this side effect with ibuzatrelvir is a meaningful patient-centric improvement.

This proactive development of a superior follow-on compound allows Pfizer to potentially transition the market to ibuzatrelvir on its own terms, mitigating future revenue loss from Paxlovid's eventual loss of exclusivity and solidifying its leadership in the oral antiviral space. The design of the Phase 3 program, particularly the dedicated trial in severely immunocompromised patients (NCT07013474), is a key component of this strategy. This patient group is among the most affected by Paxlovid's DDI issues, and a successful outcome in this trial would provide a powerful clinical rationale for establishing ibuzatrelvir as the preferred agent in this high-need population.

Preclinical Evidence Base

The decision to advance ibuzatrelvir into human clinical trials was supported by a robust body of preclinical evidence demonstrating its potent antiviral activity, oral efficacy in animal models, and favorable pharmacokinetic properties.

In Vitro Antiviral Activity

Cell-based assays were used to establish the fundamental potency and selectivity of ibuzatrelvir against its viral target and in a relevant cellular context.

  • Cellular Efficacy: In a highly relevant cell model using differentiated normal human bronchial epithelial (dNHBE) cells, which mimic the primary site of respiratory virus infection, ibuzatrelvir demonstrated potent antiviral activity against SARS-CoV-2. The half-maximal effective concentration ($EC_{50}$), a measure of the drug concentration required to inhibit viral replication by 50%, was 34 nM. The 90% maximal effective concentration ($EC_{90}$) was 70 nM. These low nanomolar values indicate high intrinsic potency at the cellular level.
  • Protease Inhibition: As detailed previously, direct enzymatic assays confirmed potent inhibition of the Mpro from SARS-CoV-2 ($IC_{50}$ = 19 nM), SARS-CoV-1 ($IC_{50}$ = 18 nM), and MERS-CoV ($IC_{50}$ = 930 nM or 65 nM, depending on the assay).
  • Selectivity: Across a panel of in vitro assays, ibuzatrelvir demonstrated a favorable off-target selectivity profile. This indicates that the compound is highly specific for its intended viral target and has a low likelihood of interacting with other host enzymes or receptors, which is a crucial predictor of a clean safety profile in humans.

In Vivo Efficacy and Pharmacokinetics (PK)

Studies in animal models were critical for demonstrating that the in vitro potency of ibuzatrelvir could translate into therapeutic efficacy when administered orally and for characterizing its absorption, distribution, metabolism, and excretion (ADME) properties.

  • Efficacy in a Mouse Model: In a mouse-adapted SARS-CoV-2 infection model, orally administered ibuzatrelvir was shown to effectively inhibit viral replication in a dose-dependent manner. Efficacy was observed at doses up to 500 mg/kg, providing essential proof-of-concept that the drug could be delivered orally and reach sufficient concentrations in target tissues to suppress the virus.
  • Pharmacokinetics in Animals: Pharmacokinetic studies were conducted in multiple species, including rats and monkeys, to assess the drug's oral bioavailability and clearance characteristics. In rats, a 7.5 mg/kg oral dose resulted in an oral bioavailability (F%) of 21.9% and a half-life ($T_{1/2}$) of 10 hours. While modest, this level of bioavailability in preclinical species was sufficient to support its development as an oral agent for humans, especially given its high potency. These preclinical PK and efficacy data provided the necessary foundation to justify the significant investment required for human clinical trials.

Table 2: Summary of Preclinical Antiviral Activity and Pharmacokinetics of Ibuzatrelvir

ParameterTarget / ModelValueSource(s)
Enzymatic Inhibition ($IC_{50}$)SARS-CoV-2 Mpro19 nM
SARS-CoV-1 Mpro18 nM
MERS-CoV Mpro65 nM - 930 nM
Cellular Antiviral ActivitySARS-CoV-2 (dNHBE cells)$EC_{50}$ = 34 nM
SARS-CoV-2 (dNHBE cells)$EC_{90}$ = 70 nM
In Vivo EfficacyMouse-adapted SARS-CoV-2 modelDose-dependent inhibition of viral replication
Pharmacokinetics (Rat)Oral Dose7.5 mg/kg
Oral Bioavailability (F%)21.9%
Half-life ($T_{1/2}$)10 hours

Clinical Development Program: From First-in-Human to Pivotal Trials

The clinical development of ibuzatrelvir has followed a logical and efficient progression from initial safety and pharmacokinetic assessments in healthy volunteers to robust dose-finding studies in patients, culminating in a large-scale, strategically designed Phase 3 program.

Phase 1 Studies

Multiple Phase 1 studies were conducted in healthy adult volunteers to establish the foundational safety, tolerability, and pharmacokinetic profile of ibuzatrelvir. Key trials in this phase include C5091013 and NCT05580003.

  • Objectives: The primary objectives of these first-in-human studies were to evaluate the safety and tolerability of single and multiple ascending doses of ibuzatrelvir and to characterize its human pharmacokinetics (i.e., how the body absorbs, distributes, metabolizes, and excretes the drug). A secondary but critical objective was to evaluate different oral formulations, including a liquid solution, a standard crystalline tablet, and a spray-dried dispersed (SDD) tablet, to select the optimal formulation for later-stage development. The studies also assessed the effect of food on the drug's absorption to provide guidance for administration.
  • Key Findings: Across these studies, ibuzatrelvir was found to be generally safe and well-tolerated at all doses tested. The most frequently reported adverse event in one study involving 12 participants was headache, which occurred in two individuals (16.7%). No serious adverse events were reported. The pharmacokinetic analyses confirmed that ibuzatrelvir is orally absorbed and enters the bloodstream, with different formulations yielding comparable total drug exposures ($AUC_{0-24}$). These data were crucial for selecting the appropriate formulation and dose levels for subsequent patient studies.

Phase 2b Dose-Ranging Study (NCT05799495 / C5091003)

Following the successful completion of Phase 1, Pfizer initiated a pivotal Phase 2b study to evaluate the antiviral efficacy and safety of ibuzatrelvir in patients with COVID-19 and to identify the optimal dose for Phase 3 development.

  • Study Design: This was a multi-center, double-blind, randomized, placebo-controlled, parallel-group trial conducted at 31 sites in the United States. The study enrolled 240 adults (aged 18 to <65 years) with confirmed, symptomatic COVID-19. A key feature of the study population was that they had no predefined risk factors for progression to severe disease, allowing for an evaluation of the drug's antiviral effect in a standard-risk population.
  • Dosing Arms: Participants were randomized in a 1:1:2:2 ratio to one of four treatment groups: placebo, ibuzatrelvir 100 mg, ibuzatrelvir 300 mg, or ibuzatrelvir 600 mg. The assigned treatment was administered orally twice daily for a duration of 5 days.
  • Primary Endpoint: The primary efficacy endpoint was the change in SARS-CoV-2 RNA viral load (VL), measured in nasopharyngeal specimens, from baseline (Day 1) to the end of treatment (Day 5).
  • Efficacy Results: The study successfully met its primary endpoint, demonstrating a clear, dose-dependent antiviral effect. All three doses of ibuzatrelvir were associated with a statistically significant and clinically meaningful greater reduction in viral load compared to placebo. The placebo-adjusted least squares mean change in viral load (in $log_{10}$ copies/mL) at Day 5 was as follows:
  • 100 mg dose: -0.7 $log_{10}$ copies/mL (80% CI: -1.1 to -0.3; P =.02)
  • 300 mg dose: -0.8 $log_{10}$ copies/mL (80% CI: -1.3 to -0.3; P =.01)
  • 600 mg dose: -1.2 $log_{10}$ copies/mL (80% CI: -1.5 to -0.8; P <.0001)
  • Safety Results: The safety profile was excellent and comparable to placebo. The incidence of adverse events was similar across all treatment groups. Importantly, no deaths or treatment-related serious adverse events occurred during the study. A key finding for tolerability was that no participants in any ibuzatrelvir group reported dysgeusia. The robust antiviral activity and acceptable safety profile from this study strongly supported the continued clinical development of ibuzatrelvir and the selection of doses for the Phase 3 program.

Table 3: Overview of the Ibuzatrelvir Clinical Trial Program

Table 4: Key Efficacy Results from the Phase 2b Study (NCT05799495)

Treatment Group (Twice Daily for 5 Days)N (Primary Analysis)Placebo-Adjusted Least Squares Mean Change in Viral Load from Baseline to Day 5 (log10​ copies/mL)80% Confidence IntervalP-value
Placebo64ReferenceN/AN/A
Ibuzatrelvir 100 mg36-0.7-1.1 to -0.30.02
Ibuzatrelvir 300 mg28-0.8-1.3 to -0.30.01
Ibuzatrelvir 600 mg71-1.2-1.5 to -0.8< 0.0001
Source:

Pivotal Phase 3 Trials (Ongoing)

Based on the strong Phase 2b results, Pfizer has initiated a comprehensive and strategically designed Phase 3 program to definitively establish the clinical efficacy and safety of ibuzatrelvir. This program consists of two distinct, large-scale, global trials targeting different high-risk patient populations. This dual-pronged strategy is highly sophisticated. The first trial (NCT06679140) is the larger, more conventional study designed to support a broad indication as a direct successor to Paxlovid. The second trial (NCT07013474) is a more focused "spearhead" study aimed at the specific patient group where Paxlovid's limitations are most acute. Success in this smaller trial could provide an early regulatory win and establish a critical market niche, de-risking the overall program and building clinical momentum.

Trial 1: High-Risk Outpatient Population (NCT06679140 / C5091017)

  • Official Title: AN INTERVENTIONAL EFFICACY AND SAFETY, PHASE 3, DOUBLE-BLIND, 2-ARM STUDY TO INVESTIGATE ORALLY ADMINISTERED IBUZATRELVIR COMPARED WITH PLACEBO IN NON-HOSPITALIZED SYMPTOMATIC ADULT AND ADOLESCENT PARTICIPANTS WITH COVID-19 WHO ARE AT HIGH RISK OF PROGRESSING TO SEVERE ILLNESS.
  • Design and Population: This is a large-scale, randomized, double-blind, placebo-controlled study expected to enroll approximately 2,330 participants. The target population is non-hospitalized adults and adolescents (12 years of age and older) with confirmed, symptomatic COVID-19 who are at high risk of progressing to severe illness due to age or underlying medical conditions.
  • Intervention: Participants are randomized 1:1 to receive either ibuzatrelvir or a matching placebo, administered orally twice daily for 5 days. Co-administration of locally available standard of care is permitted.
  • Primary Endpoint: The study has been designed to evaluate a hard clinical outcome endpoint, moving beyond the virologic surrogate used in Phase 2. The primary endpoint is the proportion of participants with COVID-19-related emergency department visits, all-cause hospitalization, or all-cause mortality through Day 28.
  • Timelines: The study began enrolling participants in late 2024, with an estimated primary completion date of December 2026.

Trial 2: Severely Immunocompromised Population (NCT07013474 / C5091018)

  • Official Title: AN INTERVENTIONAL EFFICACY AND SAFETY, PHASE 3, RANDOMIZED, DOUBLE-BLIND, 3-ARM STUDY TO INVESTIGATE IBUZATRELVIR IN ADULTS WITH SYMPTOMATIC COVID-19 WHO ARE SEVERELY IMMUNOCOMPROMISED.
  • Design and Population: This is a randomized, actively controlled, double-blind, double-dummy, 3-arm superiority study designed to enroll approximately 300 adult participants. The trial specifically targets a highly vulnerable population: adults with symptomatic COVID-19 who are severely immunocompromised due to conditions such as solid organ transplant, active hematologic malignancy, or receipt of B-cell depleting therapies.
  • Rationale and Intervention Arms: This patient population often has prolonged viral shedding and is at high risk for severe, persistent disease. They are also frequently on complex medication regimens, making them poor candidates for ritonavir-boosted therapies. The study is designed to evaluate the superiority of ibuzatrelvir-containing regimens over an active control. The three treatment arms are:
  1. Oral ibuzatrelvir + Intravenous (IV) placebo for remdesivir
  2. Oral ibuzatrelvir + IV remdesivir
  3. Oral placebo for ibuzatrelvir + IV remdesivir This design allows for the evaluation of ibuzatrelvir as both a monotherapy and in combination with the established IV antiviral remdesivir, compared against remdesivir alone.
  • Timelines: The study is actively recruiting, with an estimated primary completion date of December 2026.

Comprehensive Safety and Tolerability Profile

The safety and tolerability of ibuzatrelvir have been consistently favorable throughout its development program, from preclinical studies to late-stage clinical trials. A key feature of its safety profile is its similarity to placebo, particularly with regard to common adverse events.

Summary of Clinical Safety Data

  • Preclinical Assessment: Initial toxicological studies in animals indicated a favorable safety profile with good off-target selectivity, predicting a low risk of adverse effects in humans. The material safety data sheet for the compound classifies it as a non-hazardous substance for handling purposes.
  • Phase 1 in Healthy Volunteers: In first-in-human studies, ibuzatrelvir was generally safe and well-tolerated when administered as single or multiple doses. In a study of 12 healthy participants receiving a 300 mg dose, 50% experienced at least one medical problem. The most common of these was headache, reported by 2 participants (16.7%). No serious medical problems or deaths occurred during the study.
  • Phase 2b in COVID-19 Patients: The safety data from the dose-ranging study in 237 treated participants provided strong evidence of the drug's excellent tolerability. The overall incidence of adverse events (AEs) was low and comparable across all groups, including placebo. At least one AE was reported by 11.4% of participants in the placebo group, compared to 5.0% in the 100 mg ibuzatrelvir group, 12.8% in the 300 mg group, and 13.9% in the 600 mg group. This lack of a clear dose-dependent increase in AEs provides strong support for the drug's safety.
  • One participant in the 300 mg group discontinued treatment due to vomiting, which was considered possibly related to the study drug.
  • One participant in the 600 mg group stopped treatment due to elevated liver enzymes (ALT and AST), though it was noted that these enzymes were already high at baseline.
  • Crucially, no deaths or treatment-related serious adverse events (SAEs) occurred throughout the study's 33-day observation period.

Key Safety Advantages

The clinical data have highlighted two major safety and tolerability advantages for ibuzatrelvir when compared to the current standard-of-care oral antiviral, Paxlovid.

  • Absence of Dysgeusia: A consistent and noteworthy finding across the clinical program is the complete absence of reported cases of dysgeusia, or taste disturbance. This unpleasant side effect, often described as a metallic or bitter taste ("Paxlovid mouth"), is a very common complaint among patients taking nirmatrelvir/ritonavir and can be distressing enough to affect treatment adherence. The lack of this side effect represents a significant improvement in the patient experience.
  • Low Potential for Drug-Drug Interactions (DDIs): This is the foundational safety advantage of ibuzatrelvir. As established, its improved metabolic stability eliminates the need for ritonavir, the component of Paxlovid responsible for the vast majority of its DDI liabilities. This substantially reduces the risk of clinically significant interactions with concomitant medications, making ibuzatrelvir a potentially much safer option for patients with comorbidities who require polypharmacy.

Table 5: Consolidated Safety Profile and Adverse Events from the Phase 2b Study (NCT05799495)

Adverse Event ProfilePlacebo (N=79)Ibuzatrelvir 100 mg (N=40)Ibuzatrelvir 300 mg (N=39)Ibuzatrelvir 600 mg (N=79)
Participants with ≥1 AE, n (%)9 (11.4%)2 (5.0%)5 (12.8%)11 (13.9%)
Serious AEs, n (%)0 (0%)0 (0%)0 (0%)0 (0%)
Deaths, n (%)0 (0%)0 (0%)0 (0%)0 (0%)
Discontinuation due to AE, n (%)0 (0%)0 (0%)1 (2.6%)*1 (1.3%)**
Dysgeusia (Taste Disturbance), n (%)0 (0%)0 (0%)0 (0%)0 (0%)
Most Common AEs (≥2 participants total)*
Diarrhea, n (%)1 (1.3%)0 (0%)0 (0%)1 (1.3%)
Headache, n (%)1 (1.3%)0 (0%)1 (2.6%)0 (0%)
Nausea, n (%)0 (0%)0 (0%)1 (2.6%)1 (1.3%)
*Vomiting, possibly related to study drug. **Elevated liver enzymes (high at baseline). ***Data from plain language summary.

Regulatory Status and Global Outlook

As an investigational compound in late-stage development, ibuzatrelvir is not yet approved for marketing in any country. However, its regulatory pathway is being actively shaped by interactions with major global health authorities, and its development program is designed to support worldwide submissions.

United States (FDA)

  • Status: In the United States, ibuzatrelvir remains an investigational drug undergoing Phase 3 clinical trials. It is not yet approved by the U.S. Food and Drug Administration (FDA) for use outside of these research studies.
  • Designation: Ibuzatrelvir has been granted Fast Track designation by the FDA. This is a significant regulatory milestone. The Fast Track process is designed to facilitate the development and expedite the review of drugs intended to treat serious conditions and fill an unmet medical need. The granting of this status to ibuzatrelvir signifies that the FDA recognizes its potential to treat COVID-19 in patients for whom the current standard of care (Paxlovid) may be unsuitable due to its DDI profile. This designation allows for more frequent communication with the FDA and makes the drug eligible for Accelerated Approval and Priority Review, potentially shortening the time to market upon successful completion of Phase 3 trials.

European Union (EMA)

  • Status: Ibuzatrelvir does not currently have marketing authorization from the European Medicines Agency (EMA). However, Pfizer is clearly pursuing a coordinated global regulatory strategy that includes Europe. The pivotal Phase 3 trials (NCT06679140 and NCT07013474) have been registered with the EU Clinical Trials Information System (CTIS), as indicated by their unique EU Clinical Trial (CT) numbers (2024-517727-39-00 and 2024-517671-21-00, respectively). This dual registration in both U.S. and EU systems confirms that the clinical data package being generated is intended to support a future Marketing Authorisation Application (MAA) to the EMA.

Australia (TGA)

  • Status: The provided information contains no specific details regarding any regulatory submissions or interactions for ibuzatrelvir with the Therapeutic Goods Administration (TGA) of Australia. At this stage of development, this is not unexpected. Typically, submissions to the TGA and other regulatory bodies outside the U.S. and EU follow successful filings with the FDA and EMA. It is highly probable that Pfizer's global commercialization strategy will include Australia, with a submission to the TGA contingent upon positive Phase 3 results and approvals in major markets.

Global Outlook

The coordinated and simultaneous registration of the pivotal Phase 3 trials in both the United States (via ClinicalTrials.gov) and the European Union (via CTIS) is the hallmark of a well-resourced, top-tier global development program. This approach is designed to generate a single, comprehensive data package that can meet the stringent requirements of the world's major regulatory bodies. This strategy aims to achieve near-simultaneous market access in the largest and most influential pharmaceutical markets, thereby maximizing the drug's potential impact and commercial success upon approval. The Fast Track designation from the FDA provides a significant de-risking event, signaling regulatory alignment with the drug's development goals and potentially accelerating its path to patients.

Concluding Analysis and Future Perspectives

Ibuzatrelvir (PF-07817883) represents a significant and highly strategic evolution in the development of oral antiviral therapeutics for COVID-19. It is not merely an alternative to existing treatments but a rationally designed second-generation agent that directly addresses the most significant clinical liability of the current standard of care, Paxlovid. By achieving enhanced metabolic stability that obviates the need for ritonavir boosting, ibuzatrelvir is poised to offer a treatment option with a substantially improved safety profile, characterized by a markedly lower potential for drug-drug interactions.

Based on the comprehensive evidence gathered to date, the potential for ibuzatrelvir to succeed in its ongoing Phase 3 trials appears high. The Phase 2b study demonstrated robust, dose-dependent antiviral activity that was statistically significant and clinically meaningful. This efficacy was coupled with an excellent safety and tolerability profile that was comparable to placebo and notably free from the taste disturbances that plague Paxlovid. This strong foundation of evidence provides a compelling rationale for its advancement into pivotal trials assessing hard clinical outcomes like hospitalization and death.

Should the Phase 3 program yield positive results, ibuzatrelvir has the potential to become a new standard of care for the oral treatment of COVID-19, particularly for the large and growing population of patients with comorbidities and polypharmacy. These individuals, who are often at the highest risk for severe disease, are precisely the ones most likely to have contraindications to Paxlovid. By filling this critical unmet medical need, ibuzatrelvir could reshape COVID-19 treatment guidelines and capture a significant share of the oral antiviral market.

The next major catalysts for the program will be the primary completion and data readouts from the two pivotal Phase 3 trials, NCT06679140 and NCT07013474, which are anticipated in late 2026 and early 2027. Positive outcomes from these studies would pave the way for regulatory submissions to the FDA and EMA shortly thereafter. Beyond its immediate application for COVID-19, the demonstrated pan-coronavirus activity of ibuzatrelvir against SARS-CoV-1 and MERS-CoV underscores its value as a key asset for future pandemic preparedness, potentially providing a rapidly deployable first line of defense against novel coronavirus threats. The development of ibuzatrelvir highlights the power of modern, structure-guided medicinal chemistry to rapidly deliver refined and improved therapeutic solutions to pressing global health challenges.

Works cited

  1. Ibuzatrelvir - Wikipedia, accessed October 31, 2025, https://en.wikipedia.org/wiki/Ibuzatrelvir
  2. PF-07817883 (Ibuzatrelvir): A Second-Generation SARS-CoV-2 ..., accessed October 31, 2025, https://drughunter.com/articles/pf-07817883-ibuzatrelvir-a-second-generation-sars-cov-2-main-protease-inhibitor-w
  3. Ibuzatrelvir - New Drug Approvals, accessed October 31, 2025, https://newdrugapprovals.org/2025/06/09/ibuzatrelvir/
  4. A Structural Comparison of Oral SARS-CoV-2 Drug Candidate Ibuzatrelvir Complexed with the Main Protease (Mpro) of SARS-CoV-2 and MERS-CoV | JACS Au - ACS Publications, accessed October 31, 2025, https://pubs.acs.org/doi/10.1021/jacsau.4c00508
  5. Fixing the Achilles Heel of Pfizer's Paxlovid for COVID-19 Treatment - PMC - NIH, accessed October 31, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11284777/
  6. Virologic Response and Safety of Ibuzatrelvir, A Novel SARS-CoV-2 Antiviral, in Adults With COVID-19 - PubMed, accessed October 31, 2025, https://pubmed.ncbi.nlm.nih.gov/39486089/
  7. Clinical Study Results - Pfizer, accessed October 31, 2025, https://www.pfizer.com/sites/default/files/plsr-studies/C5091003%20Plain%20Language%20Study%20Results%20Summary%20%28PLSRS%29.pdf
  8. Pfizer readies its next-generation COVID-19 antiviral | C&EN Global ..., accessed October 31, 2025, https://pubs.acs.org/doi/10.1021/cen-10306-cover3
  9. NCT06679140 | A Study to Learn About a Study Medicine Called Ibuzatrelvir in Adult and Adolescent Patients With COVID-19 Who Are Not Hospitalized But Are at Risk For Severe Disease | ClinicalTrials.gov, accessed October 31, 2025, https://clinicaltrials.gov/study/NCT06679140
  10. NCT07013474 | A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised | ClinicalTrials.gov, accessed October 31, 2025, https://www.clinicaltrials.gov/study/NCT07013474
  11. A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised | Clinical Research Trial Listing - CenterWatch, accessed October 31, 2025, https://www.centerwatch.com/clinical-trials/listings/NCT07013474/a-study-to-learn-about-the-study-medicine-ibuzatrelvir-in-adults-with-covid-19-who-are-severely-immunocompromised?state=CA&city=West%20Hills
  12. Clinical Study Results | Pfizer, accessed October 31, 2025, https://www.pfizer.com/sites/default/files/plsr-studies/C5091013%20Plain%20Language%20Study%20Results%20Summary%20%28PLSRS%29%20Phase%201%20PK%20Primary%20Endpoint.pdf
  13. Ibuzatrelvir (PF-07817883) | Virul protease Inhibitor | MedChemExpress, accessed October 31, 2025, https://www.medchemexpress.com/ibuzatrelvir.html
  14. Ibuzatrelvir: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 31, 2025, https://go.drugbank.com/drugs/DB19451
  15. An orally bioavailable SARS-CoV-2 main protease inhibitor exhibits improved affinity and reduced sensitivity to mutations - PMC - NIH, accessed October 31, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11193659/
  16. A Structural Comparison of Oral SARS-CoV-2 Drug Candidate Ibuzatrelvir Complexed with the Main Protease (Mpro) of SARS-CoV-2 and MERS-CoV - NIH, accessed October 31, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11350714/
  17. Virologic Response and Safety of Ibuzatrelvir, A Novel SARS-CoV-2 Antiviral, in Adults With COVID-19 - PMC - NIH, accessed October 31, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11912966/
  18. A Second-Generation Oral SARS-CoV-2 Main Protease Inhibitor ..., accessed October 31, 2025, https://pubs.acs.org/doi/10.1021/acs.jmedchem.3c02469
  19. NCT05580003 | A Study to Learn Safety and Blood Levels of PF-07817883 in Healthy People | ClinicalTrials.gov, accessed October 31, 2025, https://clinicaltrials.gov/study/NCT05580003
  20. Clinical Study Results - Pfizer, accessed October 31, 2025, https://www.pfizer.com/sites/default/files/plsr-studies/C5091001%20Plain%20Language%20Study%20Results%20Summary%20%28PLSRS%29%20Phase%201%20PK%20Primary%20Endpoint.pdf
  21. A Study to Learn About a Study Medicine Called Ibuzatrelvir in Adult and Adolescent Patients With COVID-19 Who Are Not Hospitalized But Are at Risk For Severe Disease, accessed October 31, 2025, https://reaganudall.org/clinical-trial/study-learn-about-study-medicine-called-ibuzatrelvir-adult-and-adolescent-patients
  22. Study Details | NCT06679140 | A Study to Learn About a Study ..., accessed October 31, 2025, https://www.clinicaltrials.gov/study/NCT06679140?utm_source=trialradar
  23. A Study to Learn About a Study Medicine Called Ibuzatrelvir in Adult and Adolescent Patients With COVID-19 Who Are Not Hospitalized But Are at Risk For Severe Disease - Pfizer Clinical Trials, accessed October 31, 2025, https://www.pfizerclinicaltrials.com/find-a-trial/nct06679140-covid-19-sars-cov-2-infection-trial
  24. Pfizer Launched Phase 3 Trial for Ibuzatrelvir, New Oral Antiviral for COVID-19 Treatment, accessed October 31, 2025, https://atlasidp.com/pfizer-launched-phase-3-trial-for-ibuzatrelvir-new-oral-antiviral-for-covid-19-treatment/
  25. Ibuzatrelvir for Coronavirus · Recruiting Participants for Phase Phase 3 Clinical Trial 2025, accessed October 31, 2025, https://www.withpower.com/trial/ibuzatrelvir-for-coronavirus-8fc47
  26. Pfizer Launched Phase 3 Trial for Ibuzatrelvir, New Oral Antiviral for COVID-19 Treatment, accessed October 31, 2025, https://www.contagionlive.com/view/pfizer-launched-phase-3-trial-for-ibuzatrelvir-a-new-oral-antiviral-for-covid-19-treatment
  27. A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised - ClinicalTrials.Veeva, accessed October 31, 2025, https://ctv.veeva.com/study/a-study-to-learn-about-the-study-medicine-ibuzatrelvir-in-adults-with-covid-19-who-are-severely-immu
  28. A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised - ClinConnect, accessed October 31, 2025, https://clinconnect.io/trials/NCT07013474
  29. A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised - Pfizer Clinical Trials, accessed October 31, 2025, https://www.pfizerclinicaltrials.com/find-a-trial/nct07013474-covid-19-infection-trial
  30. A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised - TrialX, accessed October 31, 2025, https://www.trialx.com/clinical-trials/listings/300130/a-study-to-learn-about-the-study-medicine-ibuzatrelvir-in-adults-with-covid-19-who-are-severely-immunocompromised/
  31. ASPIRE-IC COVID-19 oral antiviral clinical trial for adults who are at increased risk of prolonged or severe illness from COVID-19 C5091018|Pfizer, accessed October 31, 2025, https://www.pfizerclinicaltrials.com/nct07013474-covid-19-trial
  32. NCT07013474 | A Study to Learn About the Study Medicine Ibuzatrelvir in Adults With COVID-19 Who Are Severely Immunocompromised | ClinicalTrials.gov, accessed October 31, 2025, https://clinicaltrials.gov/study/NCT07013474
  33. Safety Data Sheet - MedchemExpress.com, accessed October 31, 2025, https://file.medchemexpress.com/batch_PDF/HY-156654/Ibuzatrelvir-SDS-MedChemExpress.pdf
  34. ibuzatrelvir | MedPath, accessed October 31, 2025, https://trial.medpath.com/drug/283c998491dced0e/ibuzatrelvir
  35. News and events | Therapeutic Goods Administration (TGA), accessed October 31, 2025, https://www.tga.gov.au/news

Published at: October 31, 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.