MedPath

Amenamevir Advanced Drug Monograph

Published:Oct 23, 2025

Generic Name

Amenamevir

Drug Type

Small Molecule

Chemical Formula

C24H26N4O5S

CAS Number

841301-32-4

A Comprehensive Monograph on Amenamevir (DB11701)

1.0 Introduction: A New Paradigm in Anti-Herpesvirus Therapy

Amenamevir represents a significant advancement in the therapeutic landscape for infections caused by human alphaherpesviruses, specifically Varicella-Zoster Virus (VZV) and Herpes Simplex Virus (HSV). It is a first-in-class, non-nucleoside antiviral agent that functions as a helicase-primase inhibitor (HPI), a novel mechanism of action that distinguishes it from the long-standing standard of care.[1] For decades, the primary treatment for these common viral infections has relied on nucleoside analogues, such as acyclovir and its prodrug valacyclovir, which target the viral DNA polymerase.[3] While effective, these agents are associated with two key clinical challenges: the emergence of drug-resistant viral strains and the requirement for frequent daily dosing, which can impede patient adherence.[4]

The development of new antiherpetic drugs with alternative mechanisms of action has been a clinical priority, driven largely by the increasing prevalence of viral strains resistant to traditional DNA polymerase inhibitors. This issue is particularly acute in immunocompromised patient populations, where prolonged viral replication can lead to the selection of resistant mutants.[2] Amenamevir's novel mechanism directly addresses this unmet need, as it targets a different and essential step in the viral replication cycle, thereby retaining its efficacy against common acyclovir-resistant HSV isolates.[6]

Beyond overcoming resistance, Amenamevir offers a profound improvement in dosing convenience. In contrast to the multiple daily doses required for older agents like acyclovir, Amenamevir is administered once daily for the treatment of herpes zoster (shingles) and as a single, patient-initiated dose for recurrent herpes simplex.[8] This simplified regimen has significant implications for improving patient adherence and, by extension, therapeutic outcomes. The value proposition of Amenamevir is therefore a dual advantage: a novel mechanism to combat resistance coupled with a simplified dosing schedule to enhance adherence. These benefits are not independent but work synergistically to improve patient care. Marketed in Japan by Maruho Co., Ltd. under the trade name Amenalief®, Amenamevir has established itself as a pivotal new option for managing herpesvirus infections.[1]

2.0 Chemical Profile and Physicochemical Characteristics

A precise understanding of a drug's chemical and physical properties is fundamental to comprehending its pharmacological behavior. Amenamevir is a small molecule antiviral agent belonging to the phenyloxadiazole class of organic compounds.[10] Its identity is unambiguously established through a comprehensive set of chemical and regulatory identifiers, summarized in Table 1.

Table 1: Chemical and Drug Identifiers for Amenamevir

Identifier TypeValueSource(s)
Generic NameAmenamevir10
Trade NameAmenalief® (Japan)1
Synonyms / Other NamesASP-2151, ASP21511
DrugBank IDDB117011
CAS Number841301-32-41
UNII94X46KW4AE1
ATC CodeJ05AX261
IUPAC NameN-(2,6-dimethylphenyl)-N-[4-(1,2,4-oxadiazol-3-yl)anilino]-2-oxoethyl]-1,1-dioxothiane-4-carboxamide1
Chemical Formula$C_{24}H_{26}N_{4}O_{5}S$1
Average Molecular Weight482.56 g/mol1
Monoisotopic Weight482.162391127 Da10
SMILESCC1=CC=CC(C)=C1N(CC(=O)NC1=CC=C(C=C1)C1=NOC=N1)C(=O)C1CCS(=O)(=O)CC11
InChIInChI=1S/C24H26N4O5S/c1-16-4-3-5-17(2)22(16)28(24(30)19-10-12-34(31,32)13-11-19)14-21(29)26-20-8-6-18(7-9-20)23-25-15-33-27-23/h3-9,15,19H,10-14H2,1-2H3,(H,26,29)1
InChIKeyMNHNIVNAFBSLLX-UHFFFAOYSA-N1

Amenamevir's molecular structure is characterized by an $\alpha$-aminoacyl-amide backbone featuring several key functional groups that dictate its activity and properties. These include a C-terminal thiopyrane dioxide ring, an N-terminal p-1,2,4-oxadiazol-3-yl phenyl group, and a 2,6-dimethylphenyl anilide substructure.[3] The three-dimensional arrangement of these groups is critical for its function. Single-crystal X-ray diffraction studies have revealed that the bulky 2,6-dimethyl substitution on the phenyl ring introduces significant steric hindrance, forcing the anilide group to twist 180 degrees out of conjugation with the adjacent amide bond.[3] In modern drug design, such conformational constraints are often intentionally engineered to lock a molecule into a specific three-dimensional shape that maximizes binding affinity and specificity for its intended biological target, while simultaneously minimizing interactions with off-target proteins. This fixed, non-planar conformation is therefore a functionally significant feature of Amenamevir's structure, likely contributing to its high potency and selectivity.

The physicochemical properties of Amenamevir have a direct and profound impact on its clinical formulation and administration. It has a very low aqueous solubility of 0.123 mg/mL and a moderate lipophilicity, with a calculated logP value between 2.03 and 2.61.[10] This poor water solubility is the scientific basis for the significant positive food effect observed in pharmacokinetic studies, where systemic exposure (AUC) increases by approximately 90% when the drug is administered with food.[12] The lipids present in a meal likely enhance the dissolution and subsequent absorption of the lipophilic drug from the gastrointestinal tract. This direct link between a fundamental chemical property and a key clinical directive has led to the strict recommendation that Amenamevir must be taken after a meal to ensure adequate bioavailability and therapeutic efficacy.[14] Despite its low solubility, Amenamevir is compliant with Lipinski's Rule of Five, which suggests it possesses drug-like properties conducive to good oral absorption potential.[10]

3.0 Mechanism of Action: Inhibition of the Viral Helicase-Primase Complex

Amenamevir exerts its antiviral effect through a novel mechanism of action that is fundamentally different from that of traditional nucleoside analogues. Its molecular target is the viral helicase-primase complex, a multi-functional enzyme system that is essential for the replication of herpesvirus DNA in VZV, HSV-1, and HSV-2.[1] This complex is responsible for two critical, sequential steps at the very beginning of the DNA replication process:

  1. Helicase Activity: The helicase component unwinds the double-stranded viral DNA helix, separating it into two single strands that can serve as templates for replication.[2]
  2. Primase Activity: The primase component synthesizes short RNA primers on the single-stranded DNA templates. These primers are essential starting points for the viral DNA polymerase to begin synthesizing new DNA strands.[2]

Amenamevir is a non-nucleoside inhibitor that binds directly to this complex, disrupting both of its enzymatic functions.[17] The precise mechanism of inhibition is sophisticated; rather than simply blocking an active site, Amenamevir stabilizes the interaction between the helicase-primase complex and its DNA substrate.[11] This action effectively jams the replication machinery, locking the complex in a non-productive state and preventing the progression of the replication fork. By halting both DNA unwinding and primer synthesis, Amenamevir completely arrests the initiation of viral DNA replication, leading to a potent suppression of viral growth.[2] This type of "molecular glue" mechanism, which sequesters the enzyme on its substrate, can be a highly effective form of inhibition and helps to explain the drug's high in-vitro potency.[8]

This mechanism stands in stark contrast to that of nucleoside analogues like acyclovir. Acyclovir is a prodrug that must first be activated via phosphorylation by the virus-encoded enzyme thymidine kinase (TK). The resulting acyclovir triphosphate then acts as a competitive inhibitor and chain terminator for the viral DNA polymerase, the enzyme responsible for the final step of DNA elongation.[4] Amenamevir's action is entirely independent of both viral TK and DNA polymerase.[3]

This mechanistic distinction has a critical clinical implication: it is the molecular basis for Amenamevir's efficacy against the most common forms of acyclovir-resistant viruses. Resistance to acyclovir frequently arises from mutations in the viral TK gene, which render the virus incapable of activating the drug.[4] Because Amenamevir's mechanism completely bypasses the need for TK-mediated phosphorylation, it remains fully active against these TK-deficient or TK-altered viral strains. This provides a crucial therapeutic alternative for patients, particularly those who are immunocompromised, who have failed treatment with first-line nucleoside analogues.[2]

4.0 Comprehensive Pharmacological Profile

The pharmacological profile of Amenamevir is defined by its potent and selective antiviral activity (pharmacodynamics) and its distinct pattern of absorption, distribution, metabolism, and excretion (pharmacokinetics). Together, these characteristics underpin its clinical efficacy and dosing regimen.

4.1 Pharmacodynamics

Amenamevir demonstrates potent and highly selective in-vitro activity against the target alphaherpesviruses. In studies using human embryonic fibroblast cells, its 50% effective concentrations ($EC_{50}$) for inhibiting viral replication were 0.047 $\mu$M for VZV, 0.036 $\mu$M for HSV-1, and 0.028 $\mu$M for HSV-2.[17] These concentrations are often more potent than those observed for acyclovir.[20] The drug's antiviral activity is coupled with a high degree of safety at the cellular level; it is not cytotoxic to host cells at therapeutic concentrations, with a 50% cytotoxic concentration ($CC_{50}$) greater than 30 $\mu$M, indicating a wide therapeutic index.[17] A key pharmacodynamic feature is its consistent potency against both wild-type and acyclovir-resistant viral strains. Preclinical studies have repeatedly confirmed that Amenamevir inhibits the replication of acyclovir-resistant VZV and HSV with $EC_{50}$ values similar to those exhibited against acyclovir-sensitive strains, validating its utility in cases of treatment failure with older agents.[6]

4.2 Pharmacokinetics

The journey of Amenamevir through the body has been well-characterized in a series of Phase 1 studies and a human mass balance study. Its pharmacokinetic properties are summarized in Table 2 and detailed below.

Table 2: Summary of Key Pharmacokinetic Parameters of Amenamevir

ParameterValue / DescriptionKey Clinical ImplicationSource(s)
Absorption (Tmax)Rapid; median 1.0–1.5 hoursOnset of action is not limited by slow absorption.22
Food EffectAUC increases by ~90% with foodMust be administered after a meal to ensure adequate bioavailability and efficacy.12
Dose ProportionalityLess than dose-proportionalIncreasing the dose does not lead to a linear increase in exposure, influencing optimal dose selection.6
Distribution~75% plasma protein bound in humansModerate protein binding.8
MetabolismExtensive; primarily via CYP3A4/5High potential for drug-drug interactions with CYP3A4 inhibitors and inducers.23
Elimination Half-Life (t½)~8–9 hours in healthy adultsSupports a convenient once-daily dosing regimen.22
Excretion RoutePrimarily fecal (74.6%); minor renal (20.6%)No dose adjustment is required for patients with renal impairment, a major clinical advantage.22

Absorption: Following oral administration, Amenamevir is rapidly absorbed, reaching peak plasma concentrations ($C_{max}$) in approximately 1.0 to 1.5 hours.[22] A notable characteristic is its less-than-dose-proportional pharmacokinetics. Studies involving single doses from 5 mg to 2400 mg showed that increases in systemic exposure (AUC and $C_{max}$) did not keep pace with increases in dose, suggesting that a process such as gastrointestinal absorption or first-pass metabolism becomes saturated at higher doses.[6] This phenomenon of diminishing returns likely guided the selection of the 400 mg and 1200 mg doses used in clinical practice, as they represent an optimal balance on the dose-response curve. As previously noted, its absorption is significantly enhanced by food.[12]

Distribution: In humans, Amenamevir is moderately bound to plasma proteins, with a binding rate of approximately 75%.[8]

Metabolism: Amenamevir undergoes extensive metabolism before elimination. In-vitro studies using human liver microsomes have identified cytochrome P450 3A4/5 (CYP3A4/5) as the principal enzyme system responsible for its metabolism.[23] The major metabolite found circulating in human plasma is a hydroxylated form known as R5.[22]

Excretion: A human mass balance study using radiolabeled Amenamevir provided a definitive picture of its elimination pathways. Following a single 200 mg oral dose, 95.3% of the radioactivity was recovered within 168 hours. The vast majority of the dose was excreted in the feces (74.6%), with a smaller fraction appearing in the urine (20.6%).[22] Critically, less than 0.1% of the parent drug was found unchanged in the feces, confirming that its elimination is almost entirely dependent on metabolism rather than direct excretion.[22] The plasma elimination half-life in healthy volunteers is approximately 8 to 9 hours, a duration that is sufficiently long to maintain therapeutic concentrations with once-daily dosing.[22]

Pharmacokinetics in Special Populations: The pharmacokinetic profile of Amenamevir is particularly advantageous for use in special populations. In patients with varying degrees of renal impairment, including those with severe, non-dialysis-dependent disease, systemic exposure was only modestly increased.[23] This, combined with the primary fecal route of elimination, means that no dose adjustment is necessary based on renal function, a significant benefit over renally-cleared antivirals, especially given that herpes zoster disproportionately affects the elderly, who often have compromised kidney function.[26] Similarly, in patients on hemodialysis, the amount of drug removed during a dialysis session was found to be minimal, obviating the need for dose adjustments or supplemental dosing post-dialysis.[23]

5.0 Clinical Efficacy in Approved Indications

The clinical utility of Amenamevir is firmly established by robust data from pivotal Phase 3 clinical trials, which formed the basis for its regulatory approvals in Japan for two distinct indications: herpes zoster and recurrent herpes simplex.

5.1 Herpes Zoster (Shingles)

The cornerstone of the herpes zoster indication is a large, multicenter, Phase 3, randomized, double-blind study that compared Amenamevir to the established standard of care, valacyclovir.[1] The study enrolled 751 immunocompetent Japanese patients who initiated treatment within 72 hours of the onset of their zoster rash. Patients were randomized into one of three arms: Amenamevir 400 mg once daily, Amenamevir 200 mg once daily, or valacyclovir 1000 mg three times daily, all for a duration of 7 days.[20]

The primary efficacy endpoint was the proportion of patients who achieved cessation of new lesion formation by Day 4 of treatment. The results, summarized in Table 3, demonstrated that the 400 mg dose of Amenamevir was non-inferior to the standard high-dose valacyclovir regimen.

Table 3: Efficacy Outcomes from Phase 3 Herpes Zoster Trial (Amenamevir vs. Valaciclovir)

Treatment ArmPrimary Endpoint: Day 4 Cessation Proportion (%)Key Secondary Endpoint: Median Days to Complete HealingKey Secondary Endpoint: Median Days to Pain Resolution
Amenamevir 400 mg QD81.1% (197/243)No significant differenceNo significant difference
Valaciclovir 1000 mg TID75.1% (184/245)No significant differenceNo significant difference
Amenamevir 200 mg QD69.6% (172/247)No significant differenceNo significant difference
Data sourced from.20 Secondary endpoint data showed no significant differences among groups.

A formal closed testing procedure confirmed the non-inferiority of Amenamevir 400 mg to valacyclovir.[8] For secondary endpoints, including the time to complete crusting of lesions, time to complete healing, and time to resolution of zoster-associated pain, no statistically significant differences were observed among the three treatment groups.[20]

The clinical significance of this trial is profound. Achieving non-inferiority to the standard of care is a major clinical success, especially when accompanied by a significant advantage in convenience. The trial successfully demonstrated that a simple 400 mg once-daily dose of Amenamevir provides comparable efficacy to a total daily dose of 3000 mg of valacyclovir administered three times a day.[28] This provides a compelling rationale for its use as a first-line agent, particularly for patients in whom adherence to a more complex, multi-dose regimen may be a concern.

5.2 Recurrent Herpes Simplex

For recurrent herpes simplex, Amenamevir was evaluated using an innovative and patient-centric approach. The pivotal study was a Phase 3, randomized, double-blind, placebo-controlled trial that investigated a single 1200 mg dose of Amenamevir in adult immunocompetent patients with a history of recurrent genital herpes.[7]

The study was designed around a Patient-Initiated Therapy (PIT) model. Patients were provided with the study drug in advance and instructed to self-administer the single dose after a meal within 6 hours of recognizing the onset of prodromal symptoms (such as localized discomfort, burning, or itching) that precede a full outbreak.[29] This strategy is designed to maximize the benefit of antiviral therapy by ensuring treatment is initiated at the earliest possible stage of viral reactivation.

The primary endpoint was the time to healing of all genital herpes lesions. The results demonstrated a clear and statistically significant superiority of Amenamevir over placebo. In the modified intention-to-treat analysis, the median time to all lesion healing was 4.0 days in the Amenamevir group compared to 5.1 days in the placebo group (Hazard Ratio 1.60; 95% CI, 1.19–2.15; $p=0.0018$).[30]

This successful outcome led to the approval of Amenamevir for recurrent herpes simplex in Japan in 2023.[9] The single-dose PIT model represents a fundamental shift in the management of this chronic, recurrent condition. It empowers patients by removing the delays inherent in scheduling a physician visit after symptoms begin, allowing for immediate intervention. This clinical management strategy leverages the drug's potent pharmacology and favorable pharmacokinetic profile, which allows for a high single dose to be effective, thereby improving patient quality of life by shortening the duration of painful episodes and potentially aborting some outbreaks altogether.

6.0 Safety, Tolerability, and Risk Management

The safety profile of Amenamevir has been characterized through a combination of rigorous clinical trials and extensive post-marketing surveillance in Japan, where it has been used in over 1.2 million patients.[32] Overall, the drug is well-tolerated, with an adverse event profile that is generally comparable to that of valacyclovir.[20]

In the pivotal Phase 3 trial for herpes zoster, the incidence of drug-related adverse events was similar between groups: 10.0% in the Amenamevir 400 mg arm versus 12.0% in the valacyclovir arm.[28] In the Phase 3 trial for recurrent genital herpes, all treatment-emergent adverse events in both the Amenamevir and placebo groups were reported as mild in severity.[29]

The most commonly reported adverse drug reactions (ADRs) in clinical use include drug rash, headache, dizziness, diarrhea, nausea, abdominal pain, increased blood pressure, and palpitations.[14] A rare but serious cutaneous reaction, erythema multiforme, has been identified, and patients are advised to discontinue the medication and seek immediate medical attention if symptoms such as a round, spotty red rash, fever, or joint pain occur.[14]

A large-scale, real-world post-marketing surveillance study conducted in Japan, involving 3,110 patients treated for herpes zoster, provided valuable insights into the drug's safety in a broader population. The overall incidence of ADRs was low, at 0.77%.[34] This study identified four serious ADRs in four separate patients: thrombocytopenia, hyponatremia, rhabdomyolysis, and rash.[34]

A notable finding from clinical development has been the frequent observation of transient increases in specific urinary renal biomarkers, namely N-acetyl-$\beta$-D-glucosaminidase (NAG) and $\alpha$1-microglobulin.[35] While such elevations might typically raise concerns about nephrotoxicity, a detailed review by Japan's Pharmaceuticals and Medical Devices Agency (PMDA) concluded that these changes are not associated with clinically significant kidney damage or a decline in renal function.[36] This apparent paradox is likely explained by a direct pharmacological effect of the drug on renal tubular cells rather than overt toxicity. This conclusion is strongly supported by the pharmacokinetic data showing that Amenamevir is primarily cleared via a non-renal pathway, and its use does not require dose adjustment in patients with renal impairment.[8] It is crucial for clinicians to understand this distinction to avoid misinterpreting laboratory results and inappropriately discontinuing an effective therapy.

Post-marketing pharmacovigilance has also identified a potential safety signal for encephalopathy, particularly in elderly patients aged 70 years or older.[39] This type of rare but serious risk often emerges from real-world data, as clinical trial populations may be younger and have fewer comorbidities. This finding underscores the importance of ongoing safety monitoring and suggests that while Amenamevir is a valuable therapeutic option, its use in the frail elderly requires heightened vigilance for central nervous system side effects.

7.0 Drug-Drug Interaction Profile: A Clinician's Guide

Amenamevir possesses a complex, bidirectional drug-drug interaction (DDI) profile centered on the cytochrome P450 (CYP) enzyme system. It is both a substrate for and a modulator (inducer and inhibitor) of several key CYP enzymes. This necessitates a careful review of a patient's concomitant medications before initiating therapy. The interactions can be categorized by Amenamevir's role as either the "victim" of an interaction (its own metabolism is affected) or the "perpetrator" (it affects the metabolism of other drugs). These interactions are summarized in Table 4.

Table 4: Summary of Clinically Significant Drug-Drug Interactions with Amenamevir

Interaction TypeInteracting Drug/ClassEnzymeObserved EffectClinical RecommendationSource(s)
Effects of Other Drugs on Amenamevir (Victim Profile)
Strong CYP3A4 Inhibitors (e.g., ketoconazole, ritonavir, grapefruit juice)CYP3A4Increase Amenamevir exposure (AUC $\uparrow$ 2.58-fold with ketoconazole)Co-administration should be avoided due to increased risk of toxicity.14
Strong CYP3A4 Inducers (e.g., rifampicin, St. John's Wort)CYP3A4Decrease Amenamevir exposure (AUC $\downarrow$ ~58% with rifampicin)Co-administration should be avoided due to risk of therapeutic failure.14
Effects of Amenamevir on Other Drugs (Perpetrator Profile)
Sensitive CYP3A4 Substrates (e.g., midazolam)CYP3A4Amenamevir induces CYP3A4, decreasing substrate exposure (Midazolam AUC $\downarrow$ ~47%)Caution is advised; the efficacy of co-administered CYP3A4 substrates may be reduced.24
CYP2B6 Substrates (e.g., bupropion)CYP2B6Amenamevir weakly induces CYP2B6, decreasing substrate exposure (Bupropion AUC $\downarrow$ ~16%)Effect is modest; dose adjustment of the substrate is not typically required.16
CYP2C8 Substrates (e.g., montelukast)CYP2C8Amenamevir weakly inhibits CYP2C8, increasing substrate exposure (Montelukast AUC $\uparrow$ ~22%)Effect is modest; dose adjustment of the substrate is not typically required.16
CYP2C9 Substrates (e.g., (S)-warfarin)CYP2C9No significant effect observed on warfarin pharmacokinetics.No interaction is expected.16

Amenamevir as a "Victim" (Substrate of CYP3A4)

Amenamevir is primarily metabolized by CYP3A4/5.[24] Consequently, its plasma concentrations are highly susceptible to modulation by strong inhibitors or inducers of this enzyme.

  • CYP3A4 Inhibitors: Co-administration with the strong CYP3A4 inhibitor ketoconazole resulted in a 2.58-fold increase in the AUC of Amenamevir, significantly elevating systemic exposure and the potential for toxicity.[24] Patients should be counseled to avoid other strong inhibitors, including certain azole antifungals, macrolide antibiotics, HIV protease inhibitors, and grapefruit juice.[14]
  • CYP3A4 Inducers: Conversely, co-administration with the potent CYP3A4 inducer rifampicin dramatically reduced Amenamevir's AUC to just 42% of its normal value, which could lead to a loss of antiviral efficacy.[24] Patients should be advised to avoid concomitant use of strong inducers, including the herbal supplement St. John's Wort.[14]

Amenamevir as a "Perpetrator" (Inducer and Inhibitor)

Amenamevir also modulates the activity of several CYP enzymes, affecting the metabolism of other drugs.

  • CYP3A4 Induction: Amenamevir acts as a moderate inducer of CYP3A4. In a clinical study, 10 days of Amenamevir treatment decreased the exposure of the sensitive CYP3A4 substrate midazolam by approximately 47%.[24] This means that Amenamevir can reduce the efficacy of other medications that are metabolized by CYP3A4.
  • CYP2B6 Induction and CYP2C8 Inhibition: Amenamevir has a dual, modest effect on other enzymes. It weakly induces CYP2B6, causing a ~16% reduction in the exposure of bupropion, and weakly inhibits CYP2C8, causing a ~22% increase in the exposure of montelukast.[41] These effects were determined to be of minor clinical significance, and dose adjustments of co-administered substrates are generally not required.[41]
  • No Effect on CYP2C9: Amenamevir does not appear to interact with the CYP2C9 pathway, as it had no significant effect on the pharmacokinetics of the sensitive substrate (S)-warfarin.[16]

Given this complex profile, clinicians must perform a two-way check when prescribing Amenamevir. First, they must assess whether the patient is taking any strong CYP3A4 inhibitors or inducers that could alter Amenamevir levels. Second, they must review the patient's medication list for any sensitive CYP3A4 substrates whose efficacy might be compromised by Amenamevir-induced metabolism.

8.0 Dosing, Administration, and Prescribing Information

The clinical use of Amenamevir is governed by specific, evidence-based dosing and administration guidelines derived from its pivotal clinical trials and pharmacological properties. The official prescribing information for Amenalief® in Japan outlines two distinct regimens for its approved indications.

Table 5: Official Dosing Regimens for Approved Indications (Amenalief®)

IndicationDosageFrequencyDurationCritical Administration NotesSource(s)
Herpes Zoster (Shingles)400 mg (2 x 200 mg tablets)Once Daily7 daysMust be taken orally after a meal.8
Recurrent Herpes Simplex1200 mg (6 x 200 mg tablets)Single Dose1 dayMust be taken orally after a meal. Initiate within 6 hours of prodromal symptoms (Patient-Initiated Therapy).9

Key Administration and Counseling Points

Each directive for patients is a direct translation of the drug's core scientific principles.

  • Administration with Food: For both indications, it is imperative that Amenamevir be administered orally after a meal.[14] Taking the medication on an empty stomach may significantly diminish its therapeutic effect. This instruction is a direct clinical application of the pharmacokinetic data demonstrating that food increases the drug's systemic exposure by approximately 90%, which is necessary to achieve optimal therapeutic concentrations.[12]
  • Patient-Initiated Therapy (PIT) for Recurrent Herpes Simplex: For the treatment of recurrent herpes simplex, the single 1200 mg dose is intended for use as a Patient-Initiated Therapy. Patients should be counseled to take the single dose immediately upon recognizing the initial (prodromal) symptoms of a recurrence, such as localized discomfort, burning, or itching, and no later than 6 hours after symptom onset.[9] The efficacy of this regimen was established under these specific timing conditions in its pivotal clinical trial.[7]
  • Missed Doses (Herpes Zoster Regimen): If a patient misses a dose in the 7-day herpes zoster regimen, they should take it as soon as they remember. However, if it is almost time for the next scheduled dose, they should skip the missed dose and resume their regular schedule. Patients should be instructed never to take two doses at once.[14]

Contraindications and Precautions

  • Contraindications: The primary contraindication is a known history of hypersensitivity to Amenamevir or any of the excipients in the formulation.[42]
  • Warnings: No black box warnings are associated with Amenamevir in the available prescribing information.
  • Drug and Food Interactions: As detailed in Section 7.0, patients must be explicitly warned to avoid consuming grapefruit juice (a CYP3A4 inhibitor) and any health foods or supplements containing St. John's Wort (a CYP3A4 inducer) during treatment, as these can significantly alter the drug's blood levels and compromise safety or efficacy.[14]
  • Pregnancy and Breastfeeding: Women who are pregnant, may become pregnant, or are breastfeeding are advised to consult with their healthcare provider before taking Amenamevir for a recurrent herpes simplex episode.[15]

9.0 Regulatory and Developmental Landscape

The development and regulatory journey of Amenamevir highlights a strategic, Asia-centric approach to bringing a novel therapeutic to market. The drug, originally identified as ASP2151, was discovered by the Japanese pharmaceutical company Astellas Pharma Inc..[26]

Recognizing the value of specialized expertise, Astellas entered into a strategic licensing agreement in August 2012, granting Maruho Co., Ltd., a Japanese company with a deep focus on dermatology, the exclusive rights to develop and commercialize Amenamevir in Japan.[26] This type of partnership is a common and effective strategy in the pharmaceutical industry, allowing a large, discovery-focused company to leverage the specialized development and marketing capabilities of a regional or therapeutic area expert to maximize a drug's potential.

Under Maruho's stewardship, Amenamevir successfully navigated the clinical trial process, leading to its first-ever worldwide marketing authorization. In July 2017, Japan's Ministry of Health, Labour and Welfare, based on a favorable review by the Pharmaceuticals and Medical Devices Agency (PMDA), approved Amenamevir for the treatment of herpes zoster.[1] The drug was subsequently launched in Japan under the brand name Amenalief® Tab. 200mg.[26]

Following the initial approval, Maruho continued clinical development to expand the drug's utility. Based on the positive results of the Phase 3 trial in recurrent genital herpes, Maruho received approval from the PMDA for this additional indication in February 2023, solidifying Amenamevir's role in managing both acute and recurrent herpesvirus infections in Japan.[9]

As of late 2024, Amenamevir's regulatory approval remains confined to Japan. It is not currently approved by the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) and is categorized as an investigational drug in Western regulatory databases.[10] While Phase 3 trials for herpes simplex virus infections have been completed, there is no public information in the provided materials regarding submissions for a New Drug Application (NDA) to the FDA or a Marketing Authorisation Application (MAA) to the EMA.

Maruho is, however, actively pursuing a broader regional strategy. In June 2024, the company announced an exclusive licensing agreement with Hyphens Pharma to develop and commercialize Amenamevir in 10 ASEAN (Association of Southeast Asian Nations) countries.[45] This move signals a deliberate focus on Asian markets, where demographic trends, such as aging populations, may present a significant clinical need for effective herpes zoster treatments.

10.0 Conclusion: The Role of Amenamevir in Modern Antiviral Therapy

Amenamevir has emerged as a landmark achievement in anti-herpesvirus therapy, representing the clinical validation of the helicase-primase complex as a druggable target. Its introduction provides clinicians and patients with a potent, differentiated therapeutic option that directly addresses the principal limitations of older antiviral agents.

The core strengths of Amenamevir are clear and compelling. Its novel mechanism of action as a helicase-primase inhibitor provides a critical tool for circumventing the most common pathways of resistance to traditional DNA polymerase inhibitors like acyclovir.[3] This makes it an invaluable option for patients with suspected or confirmed resistance. Furthermore, its clinical efficacy is well-established; it has demonstrated non-inferiority to the standard-of-care, valacyclovir, for the treatment of herpes zoster, and superiority to placebo for recurrent genital herpes.[28] These efficacy outcomes are coupled with transformative improvements in dosing convenience: a once-daily regimen for herpes zoster and a single, patient-initiated dose for recurrent herpes simplex. These simplified regimens can significantly enhance patient adherence, a key determinant of real-world effectiveness. Finally, its predominantly non-renal clearance pathway makes it a particularly suitable choice for elderly patients and those with chronic kidney disease, a key demographic for herpes zoster.[23]

However, the clinical profile of Amenamevir is not without complexity. Its utility is moderated by a significant and bidirectional drug-drug interaction profile, primarily mediated by its role as both a substrate and an inducer of the CYP3A4 enzyme.[24] This necessitates careful medication reconciliation and may limit its use in patients on complex polypharmacy regimens. Additionally, post-marketing surveillance has raised a potential safety signal for CNS effects, such as encephalopathy, in elderly patients, warranting cautious use and heightened vigilance in this population.[39]

In final assessment, Amenamevir should not be viewed as a universal replacement for acyclovir and valacyclovir, but rather as a highly valuable and distinct therapeutic choice. Its optimal place in therapy is as a first-line agent for patients in whom adherence to multi-dose regimens is a concern, and as an essential second-line therapy for patients who have failed or are suspected to be resistant to nucleoside analogues. The decision to use Amenamevir versus a traditional agent requires a nuanced, patient-specific risk-benefit analysis, weighing its convenience and novel mechanism against its DDI potential and safety considerations in specific populations. The successful development and clinical integration of Amenamevir not only provides a new solution for an old problem but also paves the way for a new generation of HPIs that will continue to advance the management of herpesvirus infections.

Works cited

  1. Amenamevir - Wikipedia, accessed October 23, 2025, https://en.wikipedia.org/wiki/Amenamevir
  2. What is the mechanism of Amenamevir? - Patsnap Synapse, accessed October 23, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-amenamevir
  3. Amenamevir by Ugi-4CR - Green Chemistry (RSC Publishing) DOI:10.1039/D2GC04869H, accessed October 23, 2025, https://pubs.rsc.org/en/content/articlehtml/2023/gc/d2gc04869h
  4. Characteristics of Helicase-Primase Inhibitor Amenamevir-Resistant Herpes Simplex Virus | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed October 23, 2025, https://journals.asm.org/doi/10.1128/aac.00494-21
  5. Acyclovir vs. Valacyclovir: Understanding the Differences | Everlywell, accessed October 23, 2025, https://www.everlywell.com/blog/virtual-care/acyclovir-vs-valacyclovir/
  6. Pharmacokinetics and Safety of Amenamevir in Healthy Subjects: Analysis of Four Randomized Phase 1 Studies | springermedizin.de, accessed October 23, 2025, https://www.springermedizin.de/pharmacokinetics-and-safety-of-amenamevir-in-healthy-subjects-an/15219938
  7. Single-Dose, Patient-Initiated Amenamevir Therapy for Recurrent Genital Herpes: A Phase 3, Randomized, Double-Blind, Placebo-Controlled Study | Open Forum Infectious Diseases | Oxford Academic, accessed October 23, 2025, https://academic.oup.com/ofid/article/9/10/ofac494/6711549
  8. (PDF) Pharmaceuticals and Medical Device Agency approval ..., accessed October 23, 2025, https://www.researchgate.net/publication/341483325_Pharmaceuticals_and_Medical_Device_Agency_approval_summary_Amenamevir_for_the_treatment_of_herpes_zoster
  9. Maruho Receives Manufacturing and Marketing Approval for a Partial Change of the Indication and Dosage/Administration for Anti-herpes Virus Agent "Amenalief Tab. 200mg" for the Treatment of Recurrent Herpes Simplex in Japan | Information, accessed October 23, 2025, https://www.maruho.co.jp/english/information/20230224.html
  10. Amenamevir: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed October 23, 2025, https://go.drugbank.com/drugs/DB11701
  11. Amenamevir | C24H26N4O5S | CID 11397521 - PubChem, accessed October 23, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Amenamevir
  12. Pharmacokinetics and Safety of Amenamevir in Healthy Subjects: Analysis of Four Randomized Phase 1 Studies - Semantic Scholar, accessed October 23, 2025, https://www.semanticscholar.org/paper/Pharmacokinetics-and-Safety-of-Amenamevir-in-of-1-Kusawake-Keirns/54c27cb9475c40cd4b2c63269d770609feb1b515
  13. (PDF) Pharmacokinetics and Safety of Amenamevir in Healthy Subjects: Analysis of Four Randomized Phase 1 Studies - ResearchGate, accessed October 23, 2025, https://www.researchgate.net/publication/321036823_Pharmacokinetics_and_Safety_of_Amenamevir_in_Healthy_Subjects_Analysis_of_Four_Randomized_Phase_1_Studies
  14. Amenalief Tablets 200mg [herpes zoster] | Kusuri-no-Shiori(Drug Information Sheet), accessed October 23, 2025, https://www.rad-ar.or.jp/siori/english/search/result?n=46301
  15. Amenalief Tablets 200mg [recurrent herpes simplex] | Kusuri-no-Shiori(Drug Information Sheet), accessed October 23, 2025, https://www.rad-ar.or.jp/siori/english/search/result?n=46302
  16. (PDF) AMENAMEVIR: A BREAKTHROUGH IN HERPESVIRUS ..., accessed October 23, 2025, https://www.researchgate.net/publication/388051505_AMENAMEVIR_A_BREAKTHROUGH_IN_HERPESVIRUS_THERAPY
  17. Amenamevir (ASP2151, CAS Number: 841301-32-4) | Cayman Chemical, accessed October 23, 2025, https://www.caymanchem.com/product/27921/amenamevir
  18. Amenamevir | Helicase-primase inhibitor (HPI) | Axon 3465, accessed October 23, 2025, https://www.axonmedchem.com/3465-amenamevir
  19. amenamevir - Drug Central, accessed October 23, 2025, https://drugcentral.org/drugcard/5266?q=action
  20. Amenamevir, a novel helicase–primase inhibitor, for treatment of herpes zoster: A randomized, double‐blind, valaciclovir‐controlled phase 3 study - PMC, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5697646/
  21. Amenamevir (ASP2151) | HSV Inhibitor - MedchemExpress.com, accessed October 23, 2025, https://www.medchemexpress.com/Amenamevir.html
  22. (PDF) An Open‐Label, Single‐Dose, Human Mass Balance Study of Amenamevir in Healthy Male Adults - ResearchGate, accessed October 23, 2025, https://www.researchgate.net/publication/328846213_An_Open-Label_Single-Dose_Human_Mass_Balance_Study_of_Amenamevir_in_Healthy_Male_Adults
  23. Pharmacokinetics and Dialyzability of a Single Oral Dose of ..., accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7467425/
  24. Pharmacokinetic Evaluation of the Interactions of Amenamevir (ASP2151) with Ketoconazole, Rifampicin, Midazolam, and Warfarin in Healthy Adults - NIH, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5702381/
  25. Absorption, Distribution, Metabolism, and Excretion of the Novel Helicase-Primase Inhibitor, Amenamevir (ASP2151), in Rodents - PMC, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6244745/
  26. Maruho receives manufacturing and marketing approval for Anti-Herpes Virus Agent "Amenalief ® Tab. 200mg" in Japan, accessed October 23, 2025, https://www.maruho.co.jp/english/information/2017070302.html
  27. Maruho Launches Anti-Herpes Virus Agent "Amenalief ® Tab. 200mg" in Japan, accessed October 23, 2025, https://www.maruho.co.jp/english/information/2017090702.html
  28. Amenamevir, a novel helicase-primase inhibitor, for treatment of herpes zoster: A randomized, double-blind, valaciclovir-controlled phase 3 study - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/28681394/
  29. Single-Dose, Patient-Initiated Amenamevir Therapy for Recurrent Genital Herpes - Oxford Academic, accessed October 23, 2025, https://academic.oup.com/ofid/article-pdf/9/10/ofac494/46555302/ofac494.pdf
  30. Single-Dose, Patient-Initiated Amenamevir Therapy for Recurrent Genital Herpes - Semantic Scholar, accessed October 23, 2025, https://pdfs.semanticscholar.org/ecd7/4b20045384fe18784b2cfd2f6eae7430b14a.pdf
  31. Single-Dose, Patient-Initiated Amenamevir Therapy for Recurrent Genital Herpes: A Phase 3, Randomized, Double-Blind, Placebo-Controlled Study - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/36267254/
  32. Amenamevir, a Helicase-Primase Inhibitor, for the Optimal Treatment of Herpes Zoster, accessed October 23, 2025, https://www.mdpi.com/1999-4915/13/8/1547
  33. Amenamevir, a Helicase-Primase Inhibitor, for the Optimal Treatment of Herpes Zoster - PMC - PubMed Central, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8402822/
  34. Amenamevir safe and effective treatment option for patients with ..., accessed October 23, 2025, https://medicaldialogues.in/dermatology/news/amenamevir-safe-and-effective-treatment-option-for-patients-with-herpes-zoster-115509
  35. CAS 841301-32-4 Amenamevir - BOC Sciences, accessed October 23, 2025, https://www.bocsci.com/product/amenamevir-cas-841301-32-4-459784.html
  36. Pharmaceuticals and Medical Device Agency approval summary: Amenamevir for the treatment of herpes zoster - Keio University, accessed October 23, 2025, https://keio.elsevierpure.com/en/publications/pharmaceuticals-and-medical-device-agency-approval-summary-amenam
  37. Maruho Files for Additional Indication and Dosage/Administration for Anti-herpes Virus Agent "Amenalief Tab. 200mg" for the Treatment of Recurrent Herpes Simplex in Japan | Information, accessed October 23, 2025, https://www.maruho.co.jp/english/information/20211214.html
  38. Pharmaceuticals and Medical Device Agency approval summary: Amenamevir for the treatment of herpes zoster - PubMed, accessed October 23, 2025, https://pubmed.ncbi.nlm.nih.gov/32424854/
  39. Amenalief (amenamevir) News - LARVOL Sigma, accessed October 23, 2025, https://sigma.larvol.com/product.php?e1=537994&tab=newstrac
  40. pmc.ncbi.nlm.nih.gov, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5702381/#:~:text=These%20findings%20confirm%20that%20amenamevir,is%20not%20an%20inducer%20of
  41. Amenamevir: Studies of Potential CYP2C8‐ and CYP2B6‐Mediated Pharmacokinetic Interactions With Montelukast and Bupropion in Healthy Volunteers - PMC, accessed October 23, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6221039/
  42. What is Amenamevir used for? - Patsnap Synapse, accessed October 23, 2025, https://synapse.patsnap.com/article/what-is-amenamevir-used-for
  43. Amenamevir アメナメビル - New Drug Approvals, accessed October 23, 2025, https://newdrugapprovals.org/2018/03/12/amenamevir-%E3%82%A2%E3%83%A1%E3%83%8A%E3%83%A1%E3%83%93%E3%83%AB/
  44. Amenamevir | New Drug Approvals, accessed October 23, 2025, https://newdrugapprovals.org/tag/amenamevir/
  45. Maruho and Hyphens Pharma Announce Exclusive License Agreement for Anti-herpes Virus Agent (INN: Amenamevir) in 10 ASEAN Countries | Information, accessed October 23, 2025, https://www.maruho.co.jp/english/information/20240605.html

Published at: October 23, 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.