C27H23F2N3O7S
1985606-14-1
Flu caused by Influenza, Acute, uncomplicated Influenza
Baloxavir marboxil, marketed under the brand name Xofluza®, represents a significant advancement in the therapeutic armamentarium against seasonal influenza. As the first antiviral agent with a novel mechanism of action to be approved in nearly two decades, it addresses the ongoing need for effective and convenient treatments for influenza A and B infections.[1] The drug's primary distinction lies in its single-dose oral regimen, a feature that fundamentally alters the clinical management of influenza by circumventing the adherence challenges associated with multi-day therapies, such as the five-day course required for oseltamivir.[4] This single-dose administration directly addresses a common point of failure in real-world antiviral therapy, where patients may prematurely discontinue treatment upon feeling better, potentially leading to suboptimal clinical outcomes and fostering the development of viral resistance.
Developed through a collaboration between Shionogi & Co., Ltd., which discovered the compound, and the Roche Group (including Genentech in the United States), baloxavir marboxil has been globally commercialized for both the treatment of acute, uncomplicated influenza and for post-exposure prophylaxis.[2] Its therapeutic profile is characterized by a broad spectrum of activity, demonstrating efficacy against both influenza A and B viruses, including strains that have developed resistance to older neuraminidase inhibitors and certain avian strains in non-clinical studies.[4] A hallmark of its clinical effect is the rapid and profound reduction in viral load, a pharmacodynamic property that distinguishes it from previous standards of care and contributes to its public health potential in reducing viral transmission.[4]
Table 1: Drug Identification and Key Properties
Property | Details | Source(s) |
---|---|---|
Non-Proprietary Name | Baloxavir marboxil | 11 |
Proprietary/Brand Name | Xofluza® | 12 |
DrugBank ID | DB13997 | 12 |
CAS Number | 1985606-14-1 | 14 |
Drug Type | Small Molecule, Prodrug | 11 |
Chemical Formula | C27H23F2N3O7S | 12 |
Molecular Weight | 571.55 g/mol | 12 |
Therapeutic Class | Antiviral; Polymerase Acidic (PA) Endonuclease Inhibitor | 9 |
ATC Code | J05AX25 | 11 |
Baloxavir marboxil is a complex small molecule designed as a prodrug to optimize oral bioavailability. Its definitive chemical identity is established through standardized nomenclature and structural identifiers, which are crucial for research, regulatory, and database purposes.
The International Union of Pure and Applied Chemistry (IUPAC) name for the compound isbenzothiepin-11-yl]-9,12-dioxo-5-oxa-1,2,8-triazatricyclo[8.4.0.0$^{3,8}$]tetradeca-10,13-dien-11-yl]oxymethyl methyl carbonate.[11] For computational and database applications, its structure is represented by the following identifiers:
The physicochemical properties of baloxavir marboxil inform its formulation and biological behavior. It exists as a solid and has a molecular weight of 571.55 g/mol, derived from its molecular formula C27H23F2N3O7S.[12] Experimental data reveal a very low aqueous solubility of 0.015 mg/mL and a LogP value of 2.26.[11] This poor solubility is a significant barrier to effective oral absorption. The chemical design as a prodrug is a direct and necessary strategy to overcome this limitation. The "marboxil" moiety is an ester group engineered to be cleaved by endogenous esterases after absorption, releasing the pharmacologically active metabolite, baloxavir acid. This prodrug approach enhances the molecule's lipophilicity and membrane permeability, thereby improving its absorption profile and enabling effective oral administration.[11] Due to its sensitivity to air and light, which can lead to oxidative degradation, the compound requires storage under desiccating conditions at -20°C.[16]
Baloxavir marboxil is the first-in-class inhibitor of the cap-dependent endonuclease (CEN), a viral enzyme essential for influenza virus replication.[4] Its mechanism of action is fundamentally different from that of neuraminidase inhibitors (e.g., oseltamivir, zanamivir), which act at the final stage of the viral life cycle by preventing the release of newly formed virions from the host cell.[14] In contrast, baloxavir marboxil targets the very initiation of viral protein synthesis.
Upon oral administration, the prodrug baloxavir marboxil is rapidly and extensively hydrolyzed to its active form, baloxavir acid.[10] The molecular target of baloxavir acid is the polymerase acidic (PA) protein, a critical subunit of the influenza virus's RNA polymerase complex.[11] Specifically, it inhibits the endonuclease activity of this protein. This enzymatic function is responsible for a process known as "cap-snatching," whereby the virus cleaves the 5' capped ends from host cell messenger RNA (mRNA) transcripts.[12] These stolen "caps" are then used as primers to initiate the transcription of viral mRNAs by the viral RNA-dependent RNA polymerase. By selectively binding to the active site of the CEN and inhibiting this cap-snatching process, baloxavir acid effectively prevents the synthesis of viral mRNAs. This halts viral gene transcription and subsequent protein production, thereby stopping viral replication at its source.[18]
The unique mechanism of baloxavir translates into potent and rapid antiviral activity. In vitro studies have quantified its high potency in enzymatic assays, with 50% inhibitory concentration (IC50) values ranging from 1.4 to 3.1 nM for influenza A viruses and 4.5 to 8.9 nM for influenza B viruses.[11] In cell culture assays, the median 50% effective concentration (
EC50) values were similarly in the low nanomolar range for various influenza A subtypes (A/H1N1 and A/H3N2) and type B strains.[15] Importantly, this activity extends to a broad spectrum of influenza viruses, including avian strains of public health concern (H5N1, H7N9) and strains resistant to neuraminidase inhibitors, as demonstrated in non-clinical studies.[4]
In vivo preclinical studies in murine models of influenza infection confirmed these findings, showing that oral administration of baloxavir marboxil led to reduced pulmonary viral loads and increased survival rates.[11] The most compelling pharmacodynamic characteristic observed in clinical trials is the rapid and substantial reduction in viral titer in infected patients. This effect is evident within 24 hours of administration and is significantly greater in both speed and magnitude than that observed with placebo or the standard five-day course of oseltamivir.[4] For instance, in a pivotal trial, the median reduction in viral load from baseline after one day of treatment was 4.8
log10TCID50/mL in the baloxavir group, compared to 2.8 log10TCID50/mL for oseltamivir and 1.3 log10TCID50/mL for placebo.[10] This superior virologic control, even when the time to symptom alleviation is similar to oseltamivir, is a key differentiator. Since viral load is a primary driver of infectivity, a more rapid decline suggests a shorter period of infectiousness. This provides a strong mechanistic rationale for the drug's demonstrated ability to reduce influenza transmission in household settings and underscores its potential as a valuable public health tool for outbreak mitigation.[8]
The pharmacokinetic profile of baloxavir marboxil is defined by its efficient conversion to the active metabolite, baloxavir acid, which exhibits a long half-life that enables the single-dose regimen.
Baloxavir marboxil is an orally bioavailable prodrug that undergoes rapid and near-complete hydrolysis to baloxavir acid by arylacetamide deacetylase and other esterases in the gastrointestinal lumen, intestinal epithelium, liver, and blood.[11] Consequently, plasma concentrations of the parent prodrug are negligible.[27] The active metabolite, baloxavir acid, reaches its peak plasma concentration (
Tmax) approximately 4 hours after oral administration in a fasted state.[10] While co-administration with food can decrease the peak concentration (
Cmax) by approximately 48% and the total exposure (AUC) by 36%, this reduction is not considered clinically significant, and the drug may be administered with or without food.[10] This is in stark contrast to the critical instruction to avoid co-administration with products containing polyvalent cations.
Baloxavir acid is extensively bound to human serum proteins, primarily albumin, with a binding percentage of approximately 93-94%.[10] It has a large apparent volume of distribution (
Vd) of approximately 1,180 L, which indicates extensive distribution from the plasma into tissues.[10]
The biotransformation of baloxavir acid occurs primarily in the liver. The major metabolic pathway is glucuronidation, mediated by the enzyme UDP-glucuronosyltransferase 1A3 (UGT1A3). A minor pathway involves oxidation by Cytochrome P450 3A4 (CYP3A4) to form a sulfoxide metabolite.[10]
The apparent terminal elimination half-life (t1/2) of baloxavir acid is notably long, averaging 79.1 hours in adults, with some studies reporting ranges up to 99 hours.[10] This prolonged half-life is the key pharmacokinetic property that underpins the efficacy of the single-dose regimen, as it maintains therapeutic drug concentrations for several days.[27] However, this long elimination phase, characterized by a slow decline in drug concentration, may also create a prolonged window of sub-therapeutic drug levels. This condition can provide a selective pressure that favors the emergence of drug-resistant viral variants, particularly in individuals with high viral loads or compromised immune responses. Excretion of the drug and its metabolites is primarily via the feces (80.1% of a radiolabeled dose), with a smaller fraction (14.7%) eliminated in the urine.[10]
Population pharmacokinetic analyses have identified body weight and race (Asian vs. non-Asian) as the most significant covariates affecting the clearance and volume of distribution of baloxavir acid.[24] These findings provide the scientific basis for the weight-based dosing strategy used in clinical practice. The drug's pharmacokinetics are not clinically meaningfully altered by moderate hepatic impairment (Child-Pugh Class B) or mild-to-moderate renal impairment (creatinine clearance
≥50 mL/min). However, its use has not been formally studied in patients with severe hepatic or renal impairment.[9]
Table 2: Summary of Key Pharmacokinetic Parameters of Baloxavir Acid in Adults
Parameter | Value (Non-Asian) | Value (Asian) | Notes | Source(s) |
---|---|---|---|---|
Tmax (Time to Peak) | ~4 hours | ~4 hours | Time to reach maximum plasma concentration. | 10 |
Protein Binding | ~93-94% | ~93-94% | Primarily to serum albumin. | 10 |
Volume of Distribution (Vd/F) | ~1,180 L | ~647 L | Large apparent volume indicates extensive tissue distribution. | 10 |
Clearance (CL/F) | ~11.0 L/hr | ~5.5 L/hr | Apparent total clearance from plasma after oral administration. | 24 |
Terminal Half-life (t1/2) | ~79.1 hours | ~79.1 hours | Long half-life supports single-dose regimen. | 10 |
Primary Route of Elimination | Feces (~80%) | Feces (~80%) | Primarily excreted in feces, with a minor portion in urine. | 10 |
The clinical development program for baloxavir marboxil has robustly established its efficacy for both the treatment and prevention of influenza across diverse patient populations. A consistent finding across pivotal trials is that while baloxavir marboxil is generally comparable to the neuraminidase inhibitor oseltamivir in terms of symptom relief duration, it is consistently superior in its virologic effects, suggesting a primary clinical advantage in reducing disease severity and transmission potential.
The CAPSTONE-1 trial (T0831) was a Phase III, randomized, placebo- and active-controlled study in otherwise healthy patients aged 12 to 64 years with uncomplicated influenza. The trial demonstrated that a single dose of baloxavir marboxil significantly reduced the median time to alleviation of symptoms (TTAS) to 53.7 hours, compared to 80.2 hours for placebo (p<0.001). This efficacy was comparable to that of a standard five-day course of oseltamivir, which resulted in a median TTAS of 53.8 hours.[3] A key secondary finding was the significantly faster reduction in viral load with baloxavir compared to both placebo and oseltamivir, reinforcing its potent pharmacodynamic effect.[10]
The CAPSTONE-2 trial (T0832) evaluated baloxavir marboxil in patients aged 12 years and older who were at high risk for developing influenza-related complications, such as individuals with asthma, chronic lung disease, diabetes, or those aged 65 and older. In this more vulnerable population, baloxavir again demonstrated superior efficacy over placebo, reducing the median time to improvement of influenza symptoms to 73.2 hours versus 102.3 hours for placebo (p<0.001). The time to symptom improvement was similar to that observed with oseltamivir (81.0 hours).[2] The rapid and superior reduction in viral shedding was also observed in this high-risk cohort, underscoring its consistent virologic activity.[9]
The miniSTONE-2 trial (CP40563) assessed the safety and efficacy of baloxavir marboxil in otherwise healthy children aged 1 to less than 12 years. The study found that a single oral dose of baloxavir was well-tolerated and effective. The median time to alleviation of influenza signs and symptoms was similar between the baloxavir group (138.1 hours) and the oseltamivir group (150.0 hours).[23] The safety profile was the primary endpoint and was found to be comparable to oseltamivir, with gastrointestinal events such as vomiting and diarrhea being the most commonly reported adverse events in both arms.[35]
The efficacy of baloxavir marboxil as a preventive agent was established in the BLOCKSTONE trial (T0834), a Phase III study conducted in household contacts of influenza-infected individuals. Participants who received a single prophylactic dose of baloxavir had a significantly lower risk of developing clinical influenza compared to those who received a placebo. The incidence of laboratory-confirmed clinical influenza was approximately 2% in the baloxavir group versus 13-14% in the placebo group, which corresponds to an 87% reduction in the likelihood of contracting the flu after exposure.[3] These results provided the basis for its approval for post-exposure prophylaxis.
The landmark CENTERSTONE trial (NCT03969212) was the first global Phase III study to demonstrate that an antiviral treatment for a respiratory virus could reduce its transmission to others. The study met its primary endpoint, showing that treating an influenza-infected individual with a single dose of baloxavir marboxil reduced the odds of the virus spreading to untreated household members by 32% compared to placebo.[8] This finding provides direct clinical evidence for the public health benefit suggested by the drug's potent effect on viral load and positions baloxavir as a potentially important tool for controlling the spread of influenza within families and communities.
Table 3: Synopsis of Pivotal Phase III Clinical Trials
Trial Name | Population | Design | Comparators | Primary Endpoint | Key Efficacy Outcome | Key Virologic Outcome |
---|---|---|---|---|---|---|
CAPSTONE-1 | Otherwise healthy patients (≥12 years) with uncomplicated influenza | Randomized, double-blind, multicenter | Placebo, Oseltamivir | Time to Alleviation of Symptoms (TTAS) | TTAS: 53.7h vs. 80.2h for placebo (p<0.001); comparable to oseltamivir (53.8h) | Significantly faster reduction in viral load vs. placebo and oseltamivir |
CAPSTONE-2 | High-risk patients (≥12 years) with uncomplicated influenza | Randomized, double-blind, multicenter | Placebo, Oseltamivir | Time to Improvement of Symptoms | 73.2h vs. 102.3h for placebo (p<0.001); comparable to oseltamivir (81.0h) | Significantly faster reduction in viral load vs. placebo and oseltamivir |
miniSTONE-2 | Otherwise healthy children (1 to <12 years) with influenza | Randomized, double-blind, active-controlled | Oseltamivir | Safety and Tolerability | TTAS comparable to oseltamivir (138.1h vs. 150.0h) | Not reported as primary outcome |
BLOCKSTONE | Household contacts (≥5 years) of influenza patients | Randomized, double-blind, placebo-controlled | Placebo | Proportion of subjects with lab-confirmed clinical influenza | 2% incidence vs. 13% for placebo (87% risk reduction) | N/A (prophylaxis trial) |
CENTERSTONE | Influenza patients and their household contacts (≥5 years) | Randomized, double-blind, placebo-controlled | Placebo | Proportion of household contacts with lab-confirmed influenza | Reduced odds of transmission by 32% vs. placebo | N/A (transmission trial) |
The overall safety profile of baloxavir marboxil is favorable, with an incidence of adverse events in clinical trials that was generally comparable to both placebo and oseltamivir.[4] The majority of reported adverse reactions are mild and transient. However, the safety assessment must also consider rare but serious hypersensitivity reactions and the public health implications of viral resistance.
In adults and adolescents (12 years of age and older), the most common adverse reactions reported in clinical studies (occurring in ≥1% of patients) were diarrhea (3%), bronchitis (3%), nausea (2%), sinusitis (2%), and headache (1%).[40] In pediatric patients (aged 5 to less than 12 years), the most frequently reported adverse reactions (occurring in
≥5% of patients) were vomiting (5%) and diarrhea (5%).[40]
Serious adverse reactions, though rare, are a critical aspect of the drug's safety profile. Post-marketing surveillance has identified cases of serious hypersensitivity reactions, including anaphylaxis, angioedema, urticaria, and erythema multiforme.[40] Consequently, baloxavir marboxil is contraindicated in patients with a known history of hypersensitivity to the drug or any of its components.[40] If an allergic-like reaction occurs or is suspected, appropriate medical treatment should be initiated immediately. Other serious events reported in post-marketing experience include psychiatric disturbances (delirium, abnormal behavior, hallucinations) and severe gastrointestinal disorders (bloody diarrhea, melena, colitis).[42]
Key warnings and precautions for prescribers include:
Baloxavir marboxil is approved by major regulatory bodies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), for the following indications:
Administration is a single oral dose, with the dosage determined by the patient's body weight. This applies to both treatment and prophylaxis indications.[4]
The medication is available as film-coated tablets and as granules for oral suspension to accommodate different age groups and patient needs.[11]
The simplicity of the single-dose regimen is counterbalanced by a critical and significant interaction with polyvalent cations. This interaction has the potential to cause treatment failure if not properly managed, elevating the importance of patient counseling to a paramount level.
The active metabolite, baloxavir acid, has chelating properties, meaning it can bind to polyvalent cations (e.g., Ca2+, Mg2+, Fe2+, Al3+, Zn2+) in the gastrointestinal tract. This binding forms an insoluble complex that is poorly absorbed, leading to significantly reduced plasma concentrations of baloxavir and a potential loss of antiviral efficacy.[12] Because the entire therapeutic course consists of a single dose, any interference with its absorption can compromise the treatment's effectiveness entirely.
Therefore, co-administration of baloxavir marboxil with the following products must be avoided:
Concurrent administration of baloxavir marboxil with the intranasal live attenuated influenza vaccine (LAIV) has not been evaluated. However, antiviral drugs may inhibit the replication of the live vaccine virus, potentially decreasing the effectiveness of the vaccination. Therefore, the concurrent use of baloxavir and LAIV should be avoided.[32] Interactions with inactivated influenza vaccines have not been studied but are not expected to be clinically significant.
Table 4: Guidance on Managing Clinically Significant Drug and Food Interactions
Interacting Substance Class | Specific Examples | Mechanism | Clinical Recommendation |
---|---|---|---|
Dairy & Calcium-Fortified Drinks | Milk, cheese, yogurt, calcium-fortified juices or plant-based milks | Chelation of baloxavir by calcium in the GI tract, preventing absorption. | AVOID co-administration. Do not take baloxavir marboxil with these products. |
Antacids & Cationic Laxatives | Tums®, Maalox®, Milk of Magnesia®; products with aluminum, calcium, magnesium | Chelation of baloxavir by polyvalent cations, preventing absorption. | AVOID co-administration. Separate administration by several hours if use is unavoidable, though avoidance is strongly preferred. |
Mineral Supplements | Multivitamins with minerals, supplements containing iron, zinc, calcium, magnesium, selenium | Chelation of baloxavir by polyvalent cations, preventing absorption. | AVOID co-administration. Patients should pause these supplements on the day they take baloxavir marboxil. |
Live Attenuated Influenza Vaccine (LAIV) | FluMist® Quadrivalent (nasal spray vaccine) | Pharmacodynamic antagonism. The antiviral may inhibit replication of the live vaccine virus. | AVOID concurrent administration. The timing of LAIV relative to baloxavir administration should be discussed with a healthcare provider. |
The emergence of viral resistance is a significant consideration for all antiviral therapies, and baloxavir marboxil is no exception. Resistance arises from specific amino acid substitutions in the drug's target, the PA protein, which reduce the binding affinity of baloxavir acid and thus diminish its inhibitory activity.[12]
The most commonly identified resistance-conferring mutations occur at position 38 of the PA protein, where isoleucine is substituted with threonine (I38T), methionine (I38M), or phenylalanine (I38F).[21] In Phase II and III clinical trials involving adults and adolescents, the incidence of treatment-emergent resistance was observed in approximately 2% to 10% of baloxavir recipients.[21]
The incidence of resistance is notably and dramatically higher in pediatric populations. In clinical trials, the rate of virus with treatment-emergent substitutions was 16% in children aged 5 to <12 years, and it rose to 40% in children younger than 5 years.[37] This striking age-dependent effect is likely due to a confluence of factors in very young children: they often experience higher and more prolonged viral shedding, and their immature immune systems may be less efficient at clearing the virus. This combination of a larger viral pool (increasing the chance of pre-existing mutants) and a less robust immune response, when coupled with the long, declining concentration tail of a single dose of baloxavir, creates a "perfect storm" of selective pressure that allows resistant variants to emerge and dominate. This high rate of resistance is the primary reason the drug is not approved for children under 5 years of age in the U.S. The potential for community transmission of these resistant strains is a public health concern that necessitates ongoing virologic surveillance.[40]
The regulatory journey of baloxavir marboxil reflects a strategic, evidence-driven approach, beginning with approval in lower-risk populations and progressively expanding to include higher-risk groups and new indications as supporting data became available.
In the United States, the FDA granted its initial approval on October 24, 2018, for the treatment of acute, uncomplicated influenza in otherwise healthy individuals aged 12 and older.[1] This was followed by a series of key indication expansions:
In the European Union, the Committee for Medicinal Products for Human Use (CHMP) of the EMA recommended approval in November 2020, and the European Commission granted marketing authorization on January 7, 2021, for both treatment and post-exposure prophylaxis in patients aged 12 and older.[3] The indication has since been expanded to include children aged 1 year and older.[38]
The clinical development of baloxavir marboxil is ongoing. Research continues to explore its utility in other patient populations, including severely ill, hospitalized patients with influenza (e.g., the INFLUENT study, NCT06653569) and in children under the age of one, demonstrating a commitment to defining its role across the full spectrum of influenza disease.[2]
Baloxavir marboxil is a mechanistically novel, highly potent antiviral agent that represents a paradigm shift in the management of uncomplicated influenza. Its primary innovation is the single-dose oral regimen, which maximizes patient adherence and convenience—a significant advantage over multi-day therapies. Clinically, its efficacy in shortening the duration of symptoms is comparable to the established standard of care, oseltamivir. However, its defining therapeutic characteristic is its superior ability to rapidly and profoundly reduce viral load. This potent virologic effect not only contributes to clinical recovery but has also been shown to reduce the transmission of influenza within households, positioning baloxavir marboxil as a valuable tool for both individual patient care and public health.
The benefits of this convenient and effective therapy must be carefully balanced against two principal liabilities. The first is a significant drug-food interaction involving chelation with polyvalent cations, which necessitates stringent and clear patient counseling to avoid co-administration with dairy products, antacids, and mineral supplements, as a single error can compromise the entire treatment course. The second is the demonstrated potential for the selection of drug-resistant viral variants, a risk that is particularly pronounced in young children and is underscored by the drug's long pharmacokinetic half-life.
In conclusion, baloxavir marboxil is a landmark addition to the anti-influenza armamentarium. Its place in therapy is secured by its unique combination of a convenient single-dose regimen, rapid virologic control, and proven ability to reduce transmission. However, responsible stewardship of this agent is paramount. Maximizing its clinical utility while preserving its long-term effectiveness will depend on meticulous patient education regarding its specific administration requirements and vigilant surveillance for the emergence and spread of viral resistance.
Published at: September 12, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.