MedPath

cilostazol;dexborneol Advanced Drug Monograph

Published:Oct 4, 2025

Vaniprevir (MK-7009): A Comprehensive Pharmacological and Clinical Monograph

Executive Summary

Vaniprevir (MK-7009) is a macrocyclic, second-generation, direct-acting antiviral agent developed by Merck & Co. as a potent inhibitor of the Hepatitis C Virus (HCV) non-structural 3/4A (NS3/4A) protease. Designed to improve upon first-generation agents, Vaniprevir demonstrated robust antiviral activity against HCV genotype 1, the most prevalent genotype in many regions. Its mechanism involves the non-covalent, competitive, and reversible inhibition of the NS3/4A serine protease, an enzyme critical for the cleavage of the viral polyprotein and subsequent viral replication.

The clinical development program for Vaniprevir, conducted primarily in combination with the then-standard-of-care peginterferon and ribavirin (PR), consistently showed high rates of virologic response. In both treatment-naïve and treatment-experienced patients with HCV genotype 1, the addition of Vaniprevir to PR significantly increased the rates of Rapid Virologic Response (RVR) and Sustained Virologic Response (SVR) compared to PR alone, establishing its clinical efficacy.

A defining characteristic of Vaniprevir is its complex and non-linear pharmacokinetic profile. The drug exhibits greater-than-dose-proportional increases in plasma exposure, likely due to the saturation of hepatic uptake transporters (OATPs) and/or metabolic pathways (CYP3A). This results in exceptionally high concentrations within the liver, the target organ of HCV infection, which contributes to its potent efficacy. However, this same pharmacokinetic behavior creates a narrow therapeutic window and a significant potential for unpredictable overexposure.

The safety profile of Vaniprevir is marked by this pharmacokinetic complexity. While generally well-tolerated in short-term clinical studies, with the most common adverse events being gastrointestinal (diarrhea, nausea), preclinical repeat-dose toxicology studies identified the liver and gallbladder as primary target organs for toxicity. This finding is mechanistically consistent with the high and sustained drug concentrations in these tissues. Furthermore, as a substrate of CYP3A and OATP transporters, Vaniprevir possesses a high potential for clinically significant drug-drug interactions, and like other early-generation protease inhibitors, it has a relatively low barrier to the development of viral resistance, with key mutations identified at positions R155 and D168.

Ultimately, the trajectory of Vaniprevir was dictated by the rapidly evolving therapeutic landscape. It received regulatory approval in Japan in 2014 under the brand name Vanihep® for use with PR. However, its global development was discontinued as the paradigm for HCV treatment shifted decisively towards all-oral, interferon-free regimens. These newer combinations offered similar or superior efficacy with a vastly improved safety, tolerability, and drug interaction profile. Vaniprevir thus represents a scientifically successful but strategically superseded therapeutic agent—a highly effective drug for an obsolete treatment paradigm, whose legacy lies in its contribution to the proof-of-concept for protease inhibition and in highlighting the limitations that drove the development of the next generation of curative HCV therapies.

1.0 Introduction: The Evolving Landscape of Hepatitis C Therapy and the Role of Protease Inhibitors

1.1 The Hepatitis C Virus and the NS3/4A Protease Target

The Hepatitis C virus (HCV), a single-stranded RNA virus of the Flaviviridae family, is a primary cause of chronic liver disease, cirrhosis, and hepatocellular carcinoma worldwide. The viral replication cycle is a complex, multi-stage process that relies on a series of viral enzymes. Upon entry into a host hepatocyte, the viral RNA is translated into a single large polyprotein, which must be cleaved by both host and viral proteases into mature structural and non-structural (NS) proteins.

A key enzyme in this process is the NS3/4A serine protease. The NS3 protein contains the catalytic domain, but its activity is critically dependent on its non-covalent association with the NS4A protein, which acts as an essential cofactor. The NS3/4A complex is responsible for four crucial proteolytic cleavages in the non-structural region of the HCV polyprotein, liberating functional proteins such as NS4A, NS4B, NS5A, and NS5B (the RNA-dependent RNA polymerase).[1] Disruption of this proteolytic cascade effectively halts the viral replication cycle, making the NS3/4A protease an exceptionally attractive and validated target for antiviral drug development.[2] The development of direct-acting antivirals (DAAs) targeting this enzyme represented a major breakthrough in the management of chronic hepatitis C.

1.2 Emergence of Direct-Acting Antivirals: Vaniprevir's Place as a Second-Generation Inhibitor

For many years, the standard of care for chronic HCV infection consisted of combination therapy with pegylated interferon-alfa (peg-IFN) and ribavirin (RBV), collectively known as PR therapy. This regimen was associated with limited efficacy, particularly in patients with HCV genotype 1, required long treatment durations, and was burdened by a high incidence of debilitating side effects.[3]

The advent of DAAs revolutionized HCV treatment. The first class of DAAs to achieve clinical success was the NS3/4A protease inhibitors. First-generation agents, such as boceprevir and telaprevir, when added to the PR backbone, significantly improved cure rates.[4] However, these early inhibitors had limitations, including complex dosing schedules, significant drug-drug interactions, and the rapid emergence of viral resistance.

This set the stage for the development of second-generation protease inhibitors, which were designed to offer improved potency, broader genotypic coverage, a higher barrier to resistance, and more favorable pharmacokinetic profiles. Vaniprevir belongs to this second generation. It is structurally characterized as a macrocyclic inhibitor, a design strategy that constrains the molecule's conformation to enhance its binding affinity for the protease active site.[3] This structural class was a deliberate chemical strategy to overcome the limitations of earlier linear protease inhibitors. Vaniprevir's entire clinical development program was predicated on its use as an add-on to the PR backbone, representing a critical but transitional phase in HCV therapy. While it was a significant improvement over the PR-only standard of care, its reliance on interferon meant it was ultimately destined to be superseded by the next therapeutic leap: all-oral, interferon-free regimens.[4]

1.3 Development History and Investigational Overview

Vaniprevir, developed by Merck & Co. under the investigational code MK-7009, underwent a comprehensive clinical development program through Phase I, II, and III trials.[14] The primary focus of these studies was the treatment of chronic HCV genotype 1 infection, evaluating the drug's safety and efficacy in both treatment-naïve patients and those who had previously failed interferon-based therapy (treatment-experienced).[14]

The drug's development culminated in regulatory approval in Japan in 2014, where it was marketed under the brand name Vanihep®.[6] However, its development for broader global markets, including the United States and Europe, was discontinued. This decision was not a reflection of a lack of efficacy but rather a strategic response to the rapid and revolutionary shift in the HCV treatment paradigm towards interferon-free DAA combinations, which rendered interferon-dependent therapies like Vaniprevir commercially and clinically obsolete.

2.0 Chemical Identity and Physicochemical Characteristics

A precise understanding of a drug's chemical and physical properties is fundamental to interpreting its pharmacological behavior, including its absorption, distribution, metabolism, and potential for formulation challenges. Vaniprevir is a complex macrocyclic peptide mimetic whose structure dictates its biological activity and pharmacokinetic profile.

2.1 Nomenclature and Chemical Identifiers

To ensure unambiguous identification, Vaniprevir is cataloged under a variety of names and registry numbers across global databases and regulatory bodies.

  • Generic/Official Names: The internationally recognized nonproprietary name is Vaniprevir (INN, USAN, JAN). Regional linguistic variants include Vaniprévir (French) and Vaniprevirum (Latin).[14]
  • Code Names/Synonyms: During its development by Merck & Co., the compound was primarily known by the code MK-7009 or MK7009.[14]
  • Brand Name: In the Japanese market, where it received approval, it is sold under the trade name Vanihep®.[6]
  • Registry Numbers: The Chemical Abstracts Service (CAS) has assigned the number 923590-37-8.[14] Its FDA Unique Ingredient Identifier (UNII) is CV3X74AO1H.[14]
  • Database Identifiers: Key database accession numbers include DrugBank ID: DB11929, ChEMBL ID: CHEMBL4525964, and PubChem CID: 24765256.[5]

2.2 Molecular Structure and Stereochemistry

The molecular architecture of Vaniprevir is complex, featuring a large macrocyclic ring system and multiple stereocenters that are critical for its specific binding to the HCV protease active site.

  • Molecular Formula: The empirical formula for Vaniprevir is .[5]
  • Molecular Weight: The calculated average molecular weight is approximately 757.94 g/mol, with a monoisotopic mass of 757.37204952 Da.[15]
  • IUPAC Name: The systematic name according to IUPAC nomenclature is (1R,21S,24S)-21-tert-butyl-N--2-ethylcyclopropyl]-16,16-dimethyl-3,19,22-trioxo-2,18-dioxa-4,20,23-triazatetracyclo[21.2.1.1$^{4,7}^{6,11}$]heptacosa-6(11),7,9-triene-24-carboxamide.[6]
  • Structural Identifiers: Standardized chemical identifiers include the InChIKey (KUQWGLQLLVFLSM-ONAXAZCASA-N) and the SMILES string (CC[C@@H]1C[C@@]1(C(=O)NS(=O)(=O)C2CC2)NC(=O)[C@@H]3C[C@@H]4CN3C(=O)C@@HC(C)(C)C).
  • Stereochemistry: The molecule possesses five defined stereocenters, and its specific biological activity is dependent on the designated ABSOLUTE stereochemistry.

2.3 Chemical Classification and Physical Properties

Vaniprevir's physical properties are characteristic of a large, lipophilic molecule designed for oral administration and are predictive of its pharmacokinetic behavior.

  • Chemical Class: Vaniprevir is classified as a small molecule, an azamacrocyclic compound, and a cyclic peptide. Its structure contains a P2 to P4 macrocyclic constraint, a key feature of its design as a second-generation protease inhibitor.
  • Physical Appearance: It is described as a tan or white to off-white, odourless powder.
  • Solubility: The drug exhibits very poor aqueous solubility, measured at 0.0125 mg/mL, a property that often presents challenges for oral absorption and formulation. It is reported to be soluble in dimethyl sulfoxide (DMSO).
  • Melting Point: The melting point is in the range of 175-177 °C.
  • Acidity/Basicity: With a strongest acidic pKa of 3.77 and a strongest basic pKa of -3.5, Vaniprevir behaves as an acidic molecule at physiological pH, carrying a formal charge of -1.
  • Lipophilicity: The partition coefficient (logP) values are reported in the range of 3.2 to 4.72, indicating that Vaniprevir is a highly lipophilic compound.
  • Drug-likeness Properties: Vaniprevir's physicochemical properties place it in the "beyond rule-of-five" chemical space, which is common for protease inhibitors that must bind to large, complex active sites. Its high molecular weight (>500 Da) and number of hydrogen bond acceptors (9-14, depending on the calculation method) lead to violations of Lipinski's Rule of Five, as well as the Ghose Filter and Veber's Rule. This profile often correlates with challenges in oral bioavailability and metabolism but is necessary for achieving high-affinity binding to the target.

The physicochemical properties of Vaniprevir are a direct consequence of its intended function. The large, complex, and lipophilic structure is necessary for potent inhibition of the NS3/4A protease active site. However, these same properties—particularly the high molecular weight and poor aqueous solubility—are known to create challenges for oral drug delivery and often lead to complex pharmacokinetic profiles, including potential food effects and reliance on transporter-mediated disposition. The high lipophilicity is also consistent with its tendency to distribute into tissues, particularly the liver, which is a key aspect of its pharmacokinetic profile.

Table 1: Summary of Vaniprevir Identifiers and Physicochemical Properties

ParameterValueSource(s)
Generic NameVaniprevir
Brand NameVanihep® (Japan)
Code NameMK-7009
DrugBank IDDB11929
CAS Number923590-37-8
Molecular Formula
Molecular Weight757.94 g/mol
IUPAC Name(1R,21S,24S)-21-tert-butyl-N--2-ethylcyclopropyl]-16,16-dimethyl-3,19,22-trioxo-2,18-dioxa-4,20,23-triazatetracyclo[21.2.1.1$^{4,7}^{6,11}$]heptacosa-6(11),7,9-triene-24-carboxamide
Chemical ClassMacrocyclic Peptide, NS3/4A Protease Inhibitor
AppearanceTan / White to off-white powder
Water Solubility0.0125 mg/mL
Melting Point175-177 °C
pKa (Strongest Acidic)3.77
logP3.2 - 4.72

3.0 Nonclinical Pharmacology

3.1 Primary Mechanism of Action: Inhibition of HCV NS3/4A Protease

The therapeutic effect of Vaniprevir is derived from its highly specific and potent activity against the HCV NS3/4A serine protease. It functions as a selective, non-covalent, competitive, and rapidly reversible inhibitor of this viral enzyme. By binding to the active site of the protease, Vaniprevir competitively blocks access of the natural substrate, the HCV polyprotein. This inhibition prevents the necessary proteolytic processing of the polyprotein into individual, functional non-structural proteins. As these proteins are essential for the formation of the viral replication complex, Vaniprevir effectively halts viral replication and maturation. The non-covalent nature of the binding means that Vaniprevir does not form a permanent bond with the enzyme, a characteristic that differentiates it from some other classes of protease inhibitors.

3.2 In Vitro Antiviral Activity and Genotypic Coverage

The potent mechanism of action of Vaniprevir translates to strong antiviral activity in preclinical cell-based models. In the HCV replicon system, a standard in vitro model where a subgenomic portion of the HCV genome autonomously replicates within human hepatoma cells (Huh-7), Vaniprevir demonstrated potent inhibition of viral replication.

The drug exhibits low-nanomolar to sub-nanomolar potency against the NS3/4A protease enzymes from HCV genotypes 1 and 2, with specific activity confirmed against genotypes 1a and 1b. In cellular assays, the half maximal effective concentration (

) values for inhibiting HCV replication were in the low nanomolar range, with reported values as low as 0.27 nM and 0.41 nM against different viral strains in Huh-7 cells. The half maximal inhibitory concentration (

) values, which measure the concentration needed to inhibit viral replication by 50%, were also in the low nanomolar range, typically between 3 nM and 19 nM, depending on the specific assay conditions and cell line used.

3.3 Secondary and Off-Target Pharmacology

While Vaniprevir was developed as a highly selective inhibitor of the HCV NS3/4A protease, in vitro screening studies have explored its activity against other targets.

One report from post-development screening suggests that Vaniprevir may also possess inhibitory activity against the papain-like protease (PL-pro) of SARS-CoV-2, the virus responsible for COVID-19. This finding is an example of drug repurposing screening and was not part of Vaniprevir's original development program. While it suggests a structural motif within Vaniprevir may have an affinity for other viral proteases, this remains a preclinical observation without clinical validation.

One database also lists a potential inhibitory action on the Gag-Pol polyprotein of Human Immunodeficiency Virus type 1 (HIV-1). However, the pharmacological action for this target is listed as "Unknown," and this finding is not corroborated by any other primary literature or clinical data. The entire clinical development program for Vaniprevir was exclusively focused on HCV, with no trials conducted in HIV-infected populations. It is therefore highly probable that this is a database artifact, a result of a non-specific in vitro assay, or a finding with no physiological or clinical relevance. The overwhelming and consistent body of evidence firmly establishes the HCV NS3/4A protease as the sole therapeutic target of Vaniprevir.

Secondary pharmacology screening against a panel of human targets suggests potential for off-target interactions at low-micromolar concentrations, including with Cathepsin S, the Adenosine A3 receptor, KDR (VEGFR2), and the Muscarinic M1 receptor. Given that the therapeutic concentrations of Vaniprevir are in the nanomolar range, the clinical relevance of these low-affinity interactions is likely negligible.

4.0 Pharmacokinetics and Biotransformation

The pharmacokinetic (PK) profile of Vaniprevir is complex and is a defining feature of the drug, influencing both its efficacy and its safety profile. Its disposition is characterized by rapid absorption, non-linear dose-exposure relationships, and extensive concentration in the liver.

4.1 Absorption and Bioavailability

  • Rate of Absorption: Following oral administration, Vaniprevir is absorbed relatively quickly. In studies with healthy male volunteers, the time to reach maximum plasma concentration () was consistently observed to be between 1 and 3 hours post-dose.
  • Dose Proportionality: A critical aspect of Vaniprevir's PK is its non-linearity. At single oral doses above 20 mg and multiple-dose regimens above 75-100 mg twice daily, both the maximum plasma concentration () and the total drug exposure, as measured by the area under the concentration-time curve (AUC), increase in a greater-than-dose-proportional manner. This effect is pronounced; clinical data from HCV-infected patients demonstrated that a two-fold increase in the dose from 300 mg to 600 mg resulted in an approximately five-fold increase in both AUC and . This non-linear behavior is likely due to the saturation of one or more clearance mechanisms, such as hepatic uptake transporters or metabolic enzymes, at higher concentrations.
  • Food Effect: The impact of food on the absorption of Vaniprevir was evaluated in a clinical study. When a single 80 mg dose was administered with a high-fat meal, the total exposure (AUC) increased modestly (Geometric Mean Ratio of 1.22 for fed vs. fasted), while the peak concentration () decreased (GMR of 0.79). These changes were not considered to be clinically meaningful for monotherapy, suggesting that the drug could be administered without regard to meals.

4.2 Distribution

  • Hepatic Concentration: A key pharmacological feature of Vaniprevir is its preferential distribution to and high concentration within the liver, the primary site of HCV replication. Preclinical studies across multiple animal species consistently demonstrated excellent liver exposure.
  • Liver-to-Plasma Ratio: This high hepatic concentration was confirmed in studies involving HCV-infected patients. Liver tissue samples obtained via biopsy showed Vaniprevir concentrations that were substantially higher than concurrent plasma concentrations. The calculated liver-to-plasma concentration ratios were highly variable but consistently large, ranging from approximately 20-fold to as high as 280-fold. For instance, in patients receiving the 600 mg dose, the mean liver concentration at 6 hours post-dose was 169 µM, a level far exceeding what is observed in plasma.
  • Role of Transporters: The active accumulation of Vaniprevir in hepatocytes is facilitated by drug transporters. Vaniprevir is a known substrate of hepatic uptake transporters, specifically solute carrier organic anion transporters (OATPs), which actively transport the drug from the bloodstream into liver cells. The saturation of these transporters at higher drug concentrations is a primary hypothesis for the observed non-linear pharmacokinetics.

4.3 Metabolism

  • Primary Pathway: The primary route of metabolic clearance for Vaniprevir involves the cytochrome P450 (CYP) enzyme system. Specifically, it has been identified as a substrate of the CYP3A isoform. As CYP3A is the most abundant drug-metabolizing enzyme in the human liver and intestine, this pathway makes Vaniprevir susceptible to a wide range of drug-drug interactions.

4.4 Excretion and Elimination

  • Half-Life and Accumulation: At steady state, Vaniprevir has a relatively short plasma terminal elimination half-life () of 4 to 6 hours. Despite this, upon repeated twice-daily dosing, the drug accumulates in the plasma. Steady-state concentrations are generally achieved within 3 to 8 days, a period longer than would be predicted by the half-life alone. This discrepancy further supports the hypothesis of saturable distribution or elimination mechanisms. The geometric mean accumulation ratios for AUC (comparing Day 14 to Day 1) were in the range of 1.53 to 1.90.
  • Route of Elimination: The primary route of elimination for Vaniprevir and its metabolites appears to be non-renal. Studies have shown that renal excretion of the unchanged parent drug is minimal. Following a high single dose of 825 mg, only about 0.2% of the dose was recovered unchanged in the urine. This strongly implies that the drug is cleared predominantly through hepatic metabolism followed by biliary and/or fecal excretion.

The non-linear, greater-than-dose-proportional pharmacokinetics of Vaniprevir is its most significant characteristic from a clinical pharmacology perspective. This behavior has direct and opposing consequences. On one hand, the efficient, transporter-mediated uptake into the liver leads to very high drug concentrations at the target site of viral replication, which is a major contributor to the drug's potent antiviral efficacy. On the other hand, this same non-linearity creates a significant safety concern. The saturation of clearance mechanisms means that small increases in dose, or the co-administration of an interacting drug that inhibits its metabolism or transport, could lead to disproportionately large and potentially toxic increases in systemic and hepatic exposure. This narrow therapeutic window and lack of predictable dose-exposure relationship represent a major clinical management challenge and a significant liability for the drug's overall profile.

5.0 Clinical Efficacy in Chronic Hepatitis C Infection

5.1 Overview of the Clinical Trial Program

Vaniprevir was evaluated in a comprehensive clinical development program that spanned Phase I, II, and III studies. The program was designed to establish the safety, tolerability, and efficacy of Vaniprevir as a component of combination therapy for chronic HCV infection.

  • Target Population: The trials primarily enrolled patients with chronic HCV genotype 1 infection, which was the most common and difficult-to-treat genotype with interferon-based therapies. The program included studies in both treatment-naïve patients and treatment-experienced patients, a group that comprised prior relapsers (those who achieved undetectable virus on therapy but rebounded after stopping) and non-responders (those who failed to achieve a virologic response during therapy).
  • Treatment Regimen: The consistent therapeutic strategy across the pivotal trials was the addition of Vaniprevir to the then-standard-of-care, which consisted of pegylated interferon (either alfa-2a or alfa-2b) and ribavirin (PR).
  • Dosing and Duration: Vaniprevir was studied across a range of doses, typically from 100 mg to 600 mg, administered either once-daily (QD) or twice-daily (BID). The duration of Vaniprevir treatment within the triple-therapy regimen varied from as short as 4 weeks to as long as 48 weeks, depending on the study protocol and patient population.

Table 2: Summary of Key Phase II/III Clinical Trials for Vaniprevir

ClinicalTrials.gov IDPhaseStudy ObjectivePatient PopulationKey Treatment Arms (Vaniprevir Dose/Duration + Backbone)Primary EndpointKey Result SummarySource(s)
NCT00704184IIEvaluate safety and efficacy of Vaniprevir + PRTreatment-Naïve, HCV Genotype 1Vaniprevir 300 mg BID, 600 mg BID, 600 mg QD, or 800 mg QD for 28 days, all with PRRapid Virologic Response (RVR) at Week 4RVR rates were 68.8%-83.3% in Vaniprevir arms vs. 5.6% in placebo arm (p < 0.001).
NCT01370642IIIEvaluate safety and efficacy of Vaniprevir + PRTreatment-Naïve, Japanese, HCV Genotype 1Vaniprevir 300 mg BID + PR for 24 weeksSustained Virologic Response 24 weeks post-treatment (SVR24)SVR24 rate was 84.5% in the Vaniprevir arm vs. 55.1% in the control arm (PR for 48 weeks) (p < 0.001).
NCT00880763IIEvaluate safety and efficacy of Vaniprevir + PRTreatment-Experienced (Relapsers), Japanese, HCV Genotype 1Vaniprevir 100, 300, or 600 mg BID + PR for 4 weeksRVR at Week 4RVR rates were 76%-95% in Vaniprevir arms vs. 20% in placebo arm (p < 0.001). Exploratory SVR rates were 95%-100% vs. 72%.
NCT00704405IIEvaluate safety, tolerability, and efficacy of Vaniprevir + PRTreatment-Experienced, HCV Genotype 1 (cirrhotic and non-cirrhotic)Vaniprevir 300 mg QD or 600 mg BID for 24 or 48 weeks, all with PRSVR24To evaluate the safety and efficacy of different Vaniprevir regimens.

5.2 Efficacy in Treatment-Naïve Patients

In patients who had not received prior treatment for HCV, the addition of Vaniprevir to PR therapy resulted in markedly superior and faster virologic responses compared to PR alone. A key Phase II dose-ranging study (NCT00704184) randomized treatment-naïve patients to receive one of four Vaniprevir regimens (300 mg BID, 600 mg BID, 600 mg QD, or 800 mg QD) or placebo, each in combination with PR for the first 28 days. The primary endpoint, RVR (undetectable HCV RNA at week 4), was achieved by 68.8% to 83.3% of patients across the Vaniprevir arms, a stark contrast to the 5.6% rate observed in the placebo-PR arm ( for all comparisons).

This early promise was confirmed in a large Phase III trial (NCT01370642) conducted in treatment-naïve Japanese patients. In this study, a 24-week course of Vaniprevir (300 mg BID) plus PR was compared against a 48-week course of PR alone. The primary efficacy endpoint, SVR24 (the clinical definition of a cure), was achieved by 84.5% of patients in the Vaniprevir arm. This was a statistically and clinically significant improvement over the 55.1% SVR24 rate in the control arm (). Notably, the Vaniprevir regimen achieved a higher cure rate in half the total treatment duration.

5.3 Efficacy in Treatment-Experienced Patients

Vaniprevir also demonstrated significant efficacy in treatment-experienced patients, a population that is historically more difficult to cure. In a Phase II study (NCT00880763) involving Japanese patients who had previously relapsed after a full course of PR therapy, the addition of Vaniprevir (at doses of 100, 300, or 600 mg BID) for the first 4 weeks of retreatment led to RVR rates of 86%, 95%, and 76%, respectively. This was significantly higher than the 20% RVR rate in the placebo-PR control group (). While exploratory, the final SVR rates in this study were exceptionally high, reaching 95% to 100% in the Vaniprevir groups, compared to 72% for the control group.

Further studies, such as the Phase II trial NCT00704405, were specifically designed to evaluate various Vaniprevir regimens in a broad treatment-experienced population, including both prior non-responders and patients with cirrhosis. Another study, NCT01405937, focused on Japanese patients who were non-responders to previous therapy. The collective results from these trials established Vaniprevir's ability to overcome prior treatment failure and effectively treat these challenging patient populations when added to PR.

5.4 Virologic Response and Resistance

Across all clinical studies, Vaniprevir demonstrated potent and rapid antiviral activity. Even as a short-term monotherapy, treatment for one week was sufficient to produce a profound decline in viral load, with HCV RNA levels decreasing by 1.8 to 4.6  IU/mL. However, as with other early-generation DAAs, monotherapy was associated with the rapid selection of drug-resistant virus.

The resistance profile of Vaniprevir was found to be predictable. In patients who experienced virologic failure, analysis of the viral genome consistently identified resistance-associated variants (RAVs) at specific amino acid positions within the NS3 protease. The most commonly detected RAVs were at positions R155 and D168, particularly in patients infected with HCV genotype 1a. The emergence of these variants conferred reduced susceptibility to Vaniprevir and was a primary mechanism of treatment failure. The predictable nature of this resistance, however, allowed for monitoring and informed the development of next-generation inhibitors designed to be active against these variants.

The clinical data unequivocally demonstrate that Vaniprevir was a highly effective antiviral drug within the therapeutic context for which it was developed. Its ability to significantly boost cure rates over the existing standard of care, both in naïve and difficult-to-treat experienced patients, was robust and consistent. This establishes that the reasons for its limited commercialization and discontinued global development were not related to a lack of efficacy, but rather to other aspects of its profile—namely safety, pharmacokinetics, and the strategic obsolescence of the interferon-based treatment paradigm itself.

6.0 Comprehensive Safety and Toxicology Assessment

A thorough evaluation of a drug's safety profile, encompassing both nonclinical toxicology studies and clinical trial adverse event data, is essential for determining its overall risk-benefit balance. For Vaniprevir, the safety assessment reveals a profile characterized by low acute toxicity but with specific target organ concerns upon repeated exposure, which are mechanistically linked to its pharmacokinetic properties.

6.1 Preclinical Toxicology Summary

The nonclinical safety program for Vaniprevir included a standard battery of toxicology studies designed to identify potential hazards prior to and during human clinical trials.

  • Acute Toxicity: Vaniprevir demonstrated a low order of acute toxicity following oral administration. In rats, the median lethal dose () was determined to be greater than 750 mg/kg, with no adverse effects observed in acute toxicity tests.
  • Repeat-Dose Toxicity: In contrast to its low acute toxicity, studies involving prolonged or repeated dosing revealed specific target organ toxicities. The primary organs of concern identified across multiple species were the liver and gallbladder. This finding led to the official GHS classification of "H373: May cause damage to organs (gallbladder, Liver) through prolonged or repeated exposure if swallowed". In some studies, particularly at higher doses, effects were also noted in the kidney, gastrointestinal tract, heart, and stomach.
  • Genotoxicity (Mutagenicity): Vaniprevir was determined to be non-genotoxic. It yielded negative results in a comprehensive panel of assays designed to detect mutagenic or clastogenic potential. These included the in vitro bacterial reverse mutation assay (Ames test), in vitro chromosomal aberration assays, and the in vivo mouse micronucleus test.
  • Carcinogenicity: Based on long-term animal studies, Vaniprevir was not found to be carcinogenic. Carcinogenicity bioassays in rats (104 weeks) and mice (6 months) were negative for any treatment-related increase in tumors.
  • Reproductive and Developmental Toxicology: Vaniprevir did not show evidence of reproductive or developmental toxicity in preclinical studies. Fertility studies in rats showed no effects on reproductive parameters. Similarly, embryo-fetal development studies in both rats and rabbits did not reveal any teratogenic effects or adverse impacts on fetal development at the doses tested.

Table 3: Summary of Preclinical Toxicology Findings

Study TypeSpeciesKey Findings (e.g., NOAEL, Target Organs, Result)Source(s)
Acute Oral ToxicityRat> 750 mg/kg; No adverse effects observed.
Repeat-Dose Toxicity (6 Months)RatNOAEL: 120 mg/kg; LOAEL: 360 mg/kg. Target Organ: Liver.
Repeat-Dose Toxicity (9 Months)DogNOAEL: 15 mg/kg; LOAEL: 30 mg/kg. Target Organs: Liver, gallbladder.
Repeat-Dose Toxicity (90 Days)MouseNOAEL: 150 mg/kg; LOAEL: 300 mg/kg. Target Organs: Liver, Kidney, GI tract, Heart, gallbladder.
Genotoxicity (In Vitro)N/ANegative in Ames, Chromosomal Aberration, and Alkaline Elution assays.
Genotoxicity (In Vivo)MouseNegative in Micronucleus test.
CarcinogenicityRat, MouseNegative in long-term bioassays.
Reproductive/Developmental ToxicityRat, RabbitNo effects on fertility or fetal development.

6.2 Clinical Safety Profile

In human clinical trials, Vaniprevir was generally well-tolerated, particularly in short-term studies. The adverse event profile was manageable and consistent with its drug class.

  • Overall Tolerability: Phase I studies in healthy volunteers and short-term monotherapy studies in HCV patients indicated that Vaniprevir was well-tolerated. There was no discernible pattern of clinically significant laboratory abnormalities or electrocardiogram (ECG) changes.
  • Common Adverse Events (AEs): When administered, either as monotherapy or in combination with PR, the most frequently reported drug-related adverse events were gastrointestinal in nature. Diarrhea and nausea were consistently reported as the most common AEs. In some studies, vomiting was also noted, with an incidence that appeared to increase at higher doses of Vaniprevir.
  • Serious Adverse Events (SAEs): In the context of short-term and dose-ranging studies, no serious adverse events were reported, and there were no discontinuations from treatment due to AEs. In longer-term combination therapy trials, the overall incidence of AEs was generally comparable between the Vaniprevir-containing arms and the placebo-PR control arm, with the notable exception of the increased incidence of the aforementioned gastrointestinal events.

6.3 Hazard Identification and Risk Management

  • Occupational Hazards: The physical form of Vaniprevir as a fine powder presents specific occupational health and safety risks. There is a potential for a dust explosion hazard if the powder is dispersed in the air in sufficient concentrations in the presence of an ignition source. Direct contact can cause mechanical irritation to the skin and eyes. Consequently, strict handling procedures, including adequate ventilation, electrical grounding, and the use of personal protective equipment (goggles, gloves), are mandated in manufacturing and formulation settings.
  • GHS Classification: Based on the preclinical toxicology data, Vaniprevir is classified under the Globally Harmonized System (GHS) as Specific Target Organ Toxicity, Repeated Exposure (STOT RE), Category 2.

A critical analysis of the safety profile reveals a direct and concerning link between Vaniprevir's unique pharmacokinetic property of high hepatic and biliary concentration and its primary target organ toxicities. The drug's mechanism of distribution, which is so beneficial for efficacy, simultaneously creates a potential for local toxicity. The sustained high-level exposure of hepatocytes and biliary epithelial cells to Vaniprevir is the most probable cause of the liver and gallbladder findings in repeat-dose animal studies. This creates a potentially narrow therapeutic window, where the concentrations required for maximal antiviral effect may approach those that cause cumulative tissue damage over time. While the clinical trials, often of shorter duration, primarily reported mild and transient AEs, the preclinical data serves as a warning for the potential risks associated with long-term, chronic administration. This inherent risk would have been a significant consideration for regulatory agencies and a clear disadvantage compared to newer antiviral agents with wider safety margins.

7.0 Drug Resistance and Interaction Profile

The long-term success of any antiviral therapy depends not only on its initial potency but also on its resilience to viral resistance and its ability to be safely co-administered with other medications. Vaniprevir, like other second-generation protease inhibitors, has a defined resistance profile and a significant potential for drug-drug interactions (DDIs) based on its metabolic pathways.

7.1 Mechanisms of Viral Resistance and Key Mutations

The HCV RNA-dependent RNA polymerase lacks proofreading capability, leading to a high mutation rate during viral replication. This results in the continuous generation of a diverse population of viral variants, or "quasispecies," within an infected individual. Some of these variants may naturally harbor amino acid substitutions that confer reduced susceptibility to DAAs, even before treatment is initiated.

Under the selective pressure of an antiviral drug like Vaniprevir, these pre-existing resistant variants can be rapidly selected for and become the dominant viral population, leading to treatment failure. For Vaniprevir, clinical studies and in vitro experiments have identified a predictable pattern of resistance.

  • Key Resistance-Associated Variants (RAVs): The most significant RAVs that confer resistance to Vaniprevir are located at amino acid positions R155 (e.g., the R155K substitution) and D168 (e.g., D168A) within the NS3 protease domain. These mutations are particularly common in patients infected with HCV genotype 1a who experience virologic failure. Mutations at position A156 (e.g., A156T) have also been identified as conferring resistance.
  • Mechanism of Resistance: Molecular modeling and computational studies have elucidated the mechanism by which these mutations reduce the drug's effectiveness. The substitutions at these key residues alter the conformation of the protease's active site, which reduces the binding affinity of Vaniprevir. Crucially, these mutations are often located outside the binding pocket for the natural polypeptide substrate. This allows the mutant protease to escape the inhibitory effect of the drug while largely retaining its normal enzymatic function, a key factor for viral fitness and the successful emergence of resistance.

7.2 Clinically Significant Drug-Drug Interactions (DDIs)

Vaniprevir's disposition in the body is dependent on major drug metabolism and transport pathways, creating a high potential for clinically significant DDIs.

  • Metabolic Pathway (CYP3A): Vaniprevir is a substrate of the cytochrome P450 3A (CYP3A) enzyme system. CYP3A is responsible for the metabolism of over half of all marketed drugs, making it a common site of DDIs.
  • Interaction with CYP3A Inhibitors: Co-administration of Vaniprevir with strong CYP3A inhibitors (e.g., ketoconazole, itraconazole, ritonavir, clarithromycin) is predicted to significantly increase Vaniprevir plasma concentrations. This is particularly hazardous given Vaniprevir's non-linear pharmacokinetics, where a moderate increase in exposure could lead to a disproportionately large rise in concentration, elevating the risk of dose-related toxicities.
  • Interaction with CYP3A Inducers: Conversely, co-administration with strong CYP3A inducers (e.g., rifampin, carbamazepine, phenytoin, St. John's Wort) is predicted to decrease Vaniprevir plasma concentrations. This could lead to sub-therapeutic drug levels, resulting in a loss of antiviral efficacy and creating an environment that facilitates the selection and emergence of drug-resistant viral variants.
  • Transporter Pathways (OATP and P-gp):
  • OATP Substrate: Vaniprevir is a substrate of the organic anion-transporting polypeptide (OATP) family of uptake transporters, which are responsible for its active transport into hepatocytes. Co-administration with potent OATP inhibitors (e.g., cyclosporine, certain other protease inhibitors like ritonavir) could impair its hepatic uptake, leading to increased plasma concentrations and a reduced liver-to-plasma ratio.
  • P-glycoprotein (P-gp): Many protease inhibitors are also substrates or modulators of the efflux transporter P-glycoprotein (P-gp), which is present in the intestine, liver, and kidneys. While not explicitly detailed for Vaniprevir, interactions via this pathway are a common class effect. P-gp inhibitors could increase its absorption and systemic exposure, while P-gp inducers could decrease it.

The combination of a relatively low barrier to resistance and a high potential for DDIs via both CYP3A and transporter pathways makes Vaniprevir a pharmacologically complex drug. The management of a patient on Vaniprevir would require careful review of all concomitant medications to avoid interactions that could either precipitate toxicity or lead to treatment failure. This high DDI potential, coupled with the need for combination therapy to prevent resistance, presents a significant clinical management challenge and a notable disadvantage compared to newer antiviral agents with more favorable and cleaner interaction profiles.

Table 4: Potential Drug-Drug Interactions for Vaniprevir Based on Known Mechanisms

8.0 Regulatory Status and Market Context

8.1 Approval in Japan

Vaniprevir successfully navigated the regulatory process in Japan. It was granted marketing approval by the Pharmaceuticals and Medical Devices Agency (PMDA) on September 25, 2014.

  • Trade Name: In the Japanese market, it is sold under the brand name Vanihep®.
  • Indication: The approved indication is for the treatment of chronic hepatitis C in patients with genotype 1 infection. This includes patients who are treatment-naïve, as well as those who were unresponsive to or relapsed after prior interferon-based therapy. A crucial component of the indication is that Vaniprevir must be used in combination with peginterferon and ribavirin.

8.2 Global Development Status

Despite its approval in Japan, Vaniprevir did not achieve marketing authorization in other major markets and remains an investigational drug in the United States and Europe.

  • FDA and EMA Status: Vaniprevir has not been approved by the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA).
  • Clinical Trial Status: While numerous clinical trials were completed to support its development, several were also withdrawn or terminated before completion, signaling a strategic shift by the developer, Merck & Co.. Expanded access programs, which provide investigational drugs to patients outside of clinical trials, were available for a time but are no longer active, further indicating the cessation of its development program.

8.3 Comparative Analysis and Market Context

The divergent regulatory outcomes for Vaniprevir can only be understood in the context of the revolutionary changes that were occurring in the field of HCV treatment during its late-stage development. Vaniprevir was developed and clinically validated as a component of a triple-therapy regimen with peginterferon and ribavirin. This approach was a significant advancement over PR therapy alone.

However, precisely during the 2013-2015 period, when Vaniprevir was approaching regulatory submission and approval, the entire therapeutic paradigm for HCV was being rendered obsolete. The approval of sofosbuvir in late 2013, followed rapidly by other DAAs from different classes (e.g., NS5A inhibitors), enabled the creation of highly effective, all-oral, interferon-free regimens. These new combinations offered cure rates that were comparable or superior to those of Vaniprevir-based triple therapy, but with dramatically improved safety and tolerability profiles (by eliminating interferon's side effects), shorter treatment durations, and simpler dosing schedules.

Vaniprevir is a poignant example of a drug that was scientifically successful but was strategically outmaneuvered by the pace of innovation. Its approval in Japan in 2014 validated its efficacy and safety profile against the regulatory standards of the time. However, by that point, the commercial and clinical landscape in Western markets had already shifted irrevocably. The prospect of launching a new therapy that still required co-administration of interferon—with all its associated toxicities and monitoring requirements—was untenable when interferon-free alternatives were becoming available. The decision by Merck to halt broader global development was almost certainly a pragmatic business decision based on the collapsing market for interferon-based therapies. The significant investment required to pursue FDA and EMA approval, particularly in light of Vaniprevir's challenging pharmacokinetic profile and preclinical toxicity signals, could not be justified when a superior therapeutic approach had already become the new standard of care. Vaniprevir's story serves as a powerful case study in the risks of pharmaceutical research and development, where a drug can become obsolete even before it reaches the global market.

9.0 Integrated Analysis and Concluding Perspectives

9.1 Synthesis of Efficacy, Safety, and Pharmacokinetic Profile

Vaniprevir (MK-7009) stands as a well-characterized but ultimately paradoxical molecule in the history of antiviral drug development. Its profile is one of potent, targeted efficacy fundamentally undermined by a challenging combination of pharmacokinetic and safety liabilities.

The drug's design as a macrocyclic NS3/4A protease inhibitor was successful, yielding a compound with sub-nanomolar potency that, in clinical trials, translated into rapid and profound viral load reduction and high cure rates for HCV genotype 1 infection. Its ability to concentrate extensively in the liver, the site of HCV replication, is a key pharmacological strength that directly contributed to this robust efficacy.

However, this strength was inextricably linked to its greatest weaknesses. The very mechanism driving its high liver concentration—saturable, transporter-mediated uptake—also produced a non-linear and unpredictable pharmacokinetic profile. This greater-than-dose-proportional exposure created a narrow therapeutic window, where the risk of toxicity could escalate rapidly with small changes in dose or interacting factors. This risk was substantiated by preclinical toxicology studies, which identified the liver and gallbladder—the organs of highest drug concentration—as the primary targets for toxicity with repeated exposure. This created a classic pharmacological dilemma: the dose required for maximal efficacy might be uncomfortably close to the threshold for long-term organ damage. Compounding these issues were a predictable but low barrier to viral resistance and a high potential for clinically significant drug-drug interactions through the CYP3A and OATP pathways, making its clinical use complex and fraught with risk.

9.2 Vaniprevir's Legacy in the Transition to Interferon-Free Therapy

Despite its limited commercial success, Vaniprevir should not be viewed as a failure. Instead, its legacy is that of an important transitional therapy and a crucial stepping stone in the path to curing Hepatitis C. It, along with its contemporaries, provided definitive proof that potent, direct-acting inhibition of key viral enzymes could lead to viral eradication. The high SVR rates achieved with Vaniprevir-based triple therapy helped solidify the central role of protease inhibitors in HCV treatment and raised the bar for what was considered a successful clinical outcome.

Simultaneously, the limitations of Vaniprevir—its reliance on the toxic interferon backbone, its complex PK, its DDI potential, and its safety signals—starkly illuminated the remaining unmet needs. These very challenges defined the ideal target product profile for the next generation of HCV drugs: compounds with simple, linear pharmacokinetics; clean safety and DDI profiles; a high barrier to resistance; and, most importantly, the ability to be combined in an all-oral, interferon-free regimen. In this sense, Vaniprevir's development helped to both validate a therapeutic strategy and catalyze the search for its successor.

9.3 Future Research and Unanswered Questions

While the chapter on Vaniprevir for HCV treatment is largely closed, the molecule itself remains a subject of scientific interest. The in vitro finding of activity against the SARS-CoV-2 papain-like protease is an intriguing example of potential repurposing, though overcoming its inherent pharmacokinetic and toxicity hurdles for a new indication would be a formidable challenge.

Ultimately, Vaniprevir serves as a valuable historical benchmark. It represents the pinnacle of the interferon-based treatment era—a highly effective drug that was rendered obsolete not by a lack of efficacy, but by the sheer speed of scientific innovation. Its story is a powerful reminder that in modern drug development, success is defined not only by a drug's intrinsic properties but also by its timing and its place within a rapidly evolving standard of care. It stands as a testament to a specific moment in the fight against Hepatitis C, a moment that was quickly surpassed by the curative, interferon-free therapies it helped to inspire.

Works cited

  1. Exploring the Drug Resistance of HCV Protease - PMC - National Institutes of Health (NIH) |, accessed October 4, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC6237193/]
  2. Vaniprevir | CAS NO.:923590-37-8 | GlpBio, accessed October 4, 2025, [https://www.glpbio.com/research\-area/vaniprevir.html]
  3. Vaniprevir plus peginterferon alfa-2a and ribavirin in treatment-experienced Japanese patients with hepatitis C virus genotype 1 infection: a randomized phase II study - PMC, accessed October 4, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC4318982/]
  4. Single‐Dose and Multiple‐Dose Pharmacokinetics of Vaniprevir in Healthy Men - PMC, accessed October 4, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC6402189/]
  5. Compound: VANIPREVIR (CHEMBL4525964) - ChEMBL - EMBL-EBI, accessed October 4, 2025, [https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL4525964]
  6. vaniprevir | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed October 4, 2025, [https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId\=11570]
  7. Characterization of Vaniprevir, a Hepatitis C Virus NS3/4A Protease Inhibitor, in Patients With HCV Genotype 1 Infection: Safety, Antiviral Activity, Resistance, and Pharmacokinetics - PubMed, accessed October 4, 2025, [https://pubmed.ncbi.nlm.nih.gov/23747481/]
  8. Computational Study on the Drug Resistance Mechanism against ..., accessed October 4, 2025, [https://pubs.acs.org/doi/10\.1021/ci400060j]
  9. Computational study on the drug resistance mechanism against HCV NS3/4A protease inhibitors vaniprevir and MK-5172 by the combination use of molecular dynamics simulation, residue interaction network, and substrate envelope analysis - PubMed, accessed October 4, 2025, [https://pubmed.ncbi.nlm.nih.gov/23745769/]
  10. Vaniprevir (MK-7009) | NS3/4A Protease Inhibitor | MedChemExpress, accessed October 4, 2025, [https://www.medchemexpress.com/vaniprevir.html]
  11. Vaniprevir plus peginterferon alfa-2b and ribavirin in treatment-naive Japanese patients with hepatitis C virus genotype 1, accessed October 4, 2025, [https://d\-nb.info/1094744719/34]
  12. Discovery of Vaniprevir (MK-7009), a Macrocyclic Hepatitis C Virus NS3/4a Protease Inhibitor | Journal of Medicinal Chemistry - ACS Publications, accessed October 4, 2025, [https://pubs.acs.org/doi/abs/10\.1021/jm9015526]
  13. Discovery of vaniprevir (MK-7009), a macrocyclic hepatitis C virus NS3/4a protease inhibitor, accessed October 4, 2025, [https://pubmed.ncbi.nlm.nih.gov/20163176/]
  14. Vaniprevir: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 4, 2025, [https://go.drugbank.com/drugs/DB11929]
  15. Vaniprevir | C38H55N5O9S | CID 24765256 - PubChem, accessed October 4, 2025, [https://pubchem.ncbi.nlm.nih.gov/compound/Vaniprevir]
  16. Vaniprevir - Wikipedia, accessed October 4, 2025, [https://en.wikipedia.org/wiki/Vaniprevir]
  17. Vaniprevir - Drug Targets, Indications, Patents - Patsnap Synapse, accessed October 4, 2025, [https://synapse.patsnap.com/drug/a71063caa95a4d96b904ce028d6d5ab6]
  18. Vaniprevir | 923590-37-8 - ChemicalBook, accessed October 4, 2025, [https://www.chemicalbook.com/ChemicalProductProperty\_EN\_CB82509978\.htm]
  19. VANIPREVIR - gsrs, accessed October 4, 2025, [https://gsrs.ncats.nih.gov/ginas/app/ui/substances/cd652941\-eb4c\-4269\-9f47\-671b4e2b4b79]
  20. Vaniprevir | C38H55N5O9S - ChemSpider, accessed October 4, 2025, [https://www.chemspider.com/Chemical\-Structure.28475381\.html]
  21. Vaniprevir Formulation - Merck.com, accessed October 4, 2025, [https://www.merck.com/docs/product/safety\-data\-sheets/hh\-sds/Vaniprevir%20Formulation\_HH\_AU\_6N.pdf]
  22. CHEBI:136047 - vaniprevir - EMBL-EBI, accessed October 4, 2025, [https://www.ebi.ac.uk/chebi/searchId.do?chebiId\=CHEBI:136047]
  23. CHEBI:136047 - vaniprevir - EMBL-EBI, accessed October 4, 2025, [https://www.ebi.ac.uk/chebi/chebiOntology.do?chebiId\=CHEBI:136047]
  24. The Molecular Basis of Drug Resistance against Hepatitis C Virus NS3/4A Protease Inhibitors - PMC, accessed October 4, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC3406087/]
  25. Discovery of Vaniprevir (MK-7009), a Macrocyclic Hepatitis C Virus NS3/4a Protease Inhibitor | Journal of Medicinal Chemistry - ACS Publications, accessed October 4, 2025, [https://pubs.acs.org/doi/abs/10\.1021/jm9015526?\&]
  26. Vaniprevir | HCV NS3/4A protease inhibitor | Buy from Supplier AdooQ®, accessed October 4, 2025, [https://www.adooq.com/vaniprevir.html]
  27. vaniprevir (PD058863, KUQWGLQLLVFLSM-ONAXAZCASA-N) - Probes & Drugs, accessed October 4, 2025, [https://www.probes\-drugs.org/compound/PD058863/]
  28. vaniprevir - Drug Central, accessed October 4, 2025, [https://drugcentral.org/drugcard/4939]
  29. Liver-to-plasma vaniprevir (MK-7009) concentration ratios in HCV-infected patients - PMC, accessed October 4, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC4714953/]
  30. Liver-to-Plasma Vaniprevir (MK-7009) Concentration Ratios in HCV-Infected Patients, accessed October 4, 2025, [https://www.researchgate.net/publication/274643549\_Liver\-to\-plasma\_vaniprevir\_MK\-7009\_concentration\_ratios\_in\_HCV\-infected\_patients]
  31. Safety and Efficacy of Vaniprevir (MK-7009) With Pegylated Interferon (Peg-IFN) and Ribavirin (RBV) in Treatment-Experienced Hepatitis C Virus (HCV) Participants (MK-7009-009) | ClinicalTrials.gov, accessed October 4, 2025, [https://clinicaltrials.gov/study/NCT00704405]
  32. Vaniprevir with pegylated interferon alpha-2a and ribavirin in treatment-naïve patients with chronic hepatitis C: a randomized phase II study - PubMed, accessed October 4, 2025, [https://pubmed.ncbi.nlm.nih.gov/22473713/]
  33. Vaniprevir plus peginterferon alfa-2a and ribavirin in treatment ..., accessed October 4, 2025, [https://pubmed.ncbi.nlm.nih.gov/25115901/]
  34. Vaniprevir Completed Phase 2 Trials for Chronic Hepatitis C Virus (HCV) Infection Treatment | DrugBank Online, accessed October 4, 2025, [https://go.drugbank.com/drugs/DB11929/clinical\_trials?conditions\=DBCOND0057091\&phase\=2\&purpose\=treatment\&status\=completed]
  35. A Study of Vaniprevir (MK-7009) in Participants With Chronic Hepatitis C Infection After ... - ClinicalTrials.gov, accessed October 4, 2025, [https://clinicaltrials.gov/study/NCT00943761]
  36. Study Details | NCT00704184 | Safety and Efficacy of Vaniprevir (MK7009) Administered With Pegylated-Interferon and Ribavirin (MK-7009-007) | ClinicalTrials.gov, accessed October 4, 2025, [https://clinicaltrials.gov/study/NCT00704184]
  37. Study Results | NCT01405937 | Study of Vaniprevir Plus PegIntron ..., accessed October 4, 2025, [https://www.clinicaltrials.gov/study/NCT01405937?term\=vaniprevir%20japanese\&viewType\=Table\&rank\=3\&tab\=results]
  38. Hepatitis C virus protease inhibitor-resistance mutations: Our experience and review, accessed October 4, 2025, [https://www.researchgate.net/publication/259499934\_Hepatitis\_C\_virus\_protease\_inhibitor\-resistance\_mutations\_Our\_experience\_and\_review]
  39. Vaniprevir Formulation | Safety Data Sheet | Merck.com, accessed October 4, 2025, [https://www.merck.com/docs/product/safety\-data\-sheets/hh\-sds/Vaniprevir%20Formulation\_HH\_CA\_EN.pdf]
  40. Vaniprevir Formulation - MSD, accessed October 4, 2025, [https://www.msd.com/docs/product/safety\-data\-sheets/hh\-sds/Vaniprevir%20Formulation\_HH\_JP\_6N.pdf]
  41. Unraveling CYP3A Gut vs Liver Drug Metabolism and P-glycoprotein Activity | Celerion, accessed October 4, 2025, [https://www.celerion.com/resource/unraveling\-cyp3a\-gut\-vs\-liver\-drug\-metabolism\-and\-p\-glycoprotein\-activity]
  42. Drug-Drug Interactions with the NS3/4A Protease Inhibitor Simeprevir - ResearchGate, accessed October 4, 2025, [https://www.researchgate.net/publication/281710711\_Drug\-Drug\_Interactions\_with\_the\_NS34A\_Protease\_Inhibitor\_Simeprevir]
  43. Toxicity and risks from drug-to-drug interactions of new antivirals for chronic hepatitis C - European Review for Medical and Pharmacological Sciences, accessed October 4, 2025, [https://www.europeanreview.org/wp/wp\-content/uploads/102\-111\-Toxicity\-and\-risks\-from\-drug\-to\-drug\-interactions\-of\-new\-antivirals\-for\-chronic\-hepatitis\-C.pdf]
  44. Examples of Major Drug-Drug Interactions with Nirmatrelvir-ritonavir and Co-administration - CMAJ, accessed October 4, 2025, [https://www.cmaj.ca/content/cmaj/suppl/2022/02/03/cmaj.220081\.DC1/220081\-five\-1\-at.pdf]
  45. Drug–Drug Interactions of Newly Approved Direct-Acting Antiviral Agents in Patients with Hepatitis C - Dove Medical Press, accessed October 4, 2025, [https://www.dovepress.com/drugndashdrug\-interactions\-of\-newly\-approved\-direct\-acting\-antiviral\-a\-peer\-reviewed\-fulltext\-article\-IJGM]
  46. Drug–drug interactions during antiviral therapy for chronic hepatitis C - PMC, accessed October 4, 2025, [https://pmc.ncbi.nlm.nih.gov/articles/PMC4634866/]
  47. P-Glycoprotein - FPnotebook, accessed October 4, 2025, [https://fpnotebook.com/Pharm/Metabolism/PGlycprtn.htm]
  48. P-glycoprotein and its role in drug-drug interactions - Australian Prescriber, accessed October 4, 2025, [https://australianprescriber.tg.org.au/articles/p\-glycoprotein\-and\-its\-role\-in\-drug\-drug\-interactions.html]
  49. NCT00518622 | Study the Safety and Effectiveness of MK7009 in Hepatitis C Infected Patients (MK-7009-004)(COMPLETED) | ClinicalTrials.gov, accessed October 4, 2025, [https://clinicaltrials.gov/study/NCT00518622?term\=AREA%5BBasicSearch%5D(vaniprevir)\&rank\=8]
  50. Maviret | European Medicines Agency (EMA), accessed October 4, 2025, [https://www.ema.europa.eu/en/medicines/human/EPAR/maviret]
  51. EMA Approves WINREVAIR (Sotatercept) for Treatment of PAH - PHA Europe, accessed October 4, 2025, [https://www.phaeurope.org/news/ema\-approves\-winrevair\-sotatercept\-for\-treatment\-of\-pah/]
  52. Merck Receives European Commission Approval for WINREVAIR™ (sotatercept) in Combination With Other Pulmonary Arterial Hypertension (PAH) Therapies, for the Treatment of PAH in Adult Patients With Functional Class II-III, accessed October 4, 2025, [https://www.merck.com/news/merck\-receives\-european\-commission\-approval\-for\-winrevair\-sotatercept\-in\-combination\-with\-other\-pulmonary\-arterial\-hypertension\-pah\-therapies\-for\-the\-treatment\-of\-pah\-in\-adult\-patients\-w/]
  53. European Medicines Agency (EMA), accessed October 4, 2025, [https://www.ema.europa.eu/en/medicines]
  54. Favipiravir - Wikipedia, accessed October 4, 2025, [https://en.wikipedia.org/wiki/Favipiravir]
  55. What is Favipiravir? FDA Approves COVID Prevention Study in US Nursing Homes, accessed October 4, 2025, [https://www.contagionlive.com/view/favipiravir\-fda\-approves\-covid\-prevention\-study\-us\-nursing\-homes]
  56. FDA Clears Appili Therapeutics to Expand its Phase 2 Clinical Trial of Favipiravir for the Potential Prevention of COVID-19 at U.S. Long-Term Care Facilities - BioSpace, accessed October 4, 2025, [https://www.biospace.com/fda\-clears\-appili\-therapeutics\-to\-expand\-its\-phase\-2\-clinical\-trial\-of\-favipiravir\-for\-the\-potential\-prevention\-of\-covid\-19\-at\-u\-s\-long\-term\-care\-facilities]
  57. FDA Approves First Treatment for COVID-19, accessed October 4, 2025, [https://www.fda.gov/news\-events/press\-announcements/fda\-approves\-first\-treatment\-covid\-19]
  58. Marketing authorisation | European Medicines Agency (EMA), accessed October 4, 2025, [https://www.ema.europa.eu/en/human\-regulatory\-overview/marketing\-authorisation]
  59. European Medicines Agency Validates Gileads Marketing Authorization Application and EU Medicines for All Application for Twice Yearly Lenacapavir for HIV Prevention - Gilead Sciences, accessed October 4, 2025, [https://www.gilead.com/news/news\-details/2025/european\-medicines\-agency\-validates\-gileads\-marketing\-authorization\-application\-and\-eu\-medicines\-for\-all\-application\-for\-twice\-yearly\-lenacapavir\-for\-hiv\-prevention]
  60. EMA grants marketing authorisation to rare disease therapies – EJP RD, accessed October 4, 2025, [https://www.ejprarediseases.org/ema\-grants\-marketing\-authorisation\-to\-rare\-disease\-therapies/]
  61. Study Details | NCT01405560 | Vaniprevir Plus PegIntron®/Ribavirin in Japanese Participants With Chronic Hepatitis C Who Are Non-responders to Previous Treatment (MK-7009-045) | ClinicalTrials.gov, accessed October 4, 2025, [https://clinicaltrials.gov/study/NCT01405560]

Published at: October 4, 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.