MedPath

Gefapixant Advanced Drug Monograph

Published:Aug 26, 2025

Generic Name

Gefapixant

Brand Names

Lyfnua

Drug Type

Small Molecule

Chemical Formula

C14H19N5O4S

CAS Number

1015787-98-0

Associated Conditions

Refractory Chronic Cough, Unexplained Chronic Cough

Gefapixant: A Comprehensive Monograph on a First-in-Class P2X3 Antagonist for Chronic Cough

I. Executive Summary

Gefapixant, marketed under the brand name Lyfnua, is a first-in-class, orally administered, selective P2X3 receptor antagonist developed for the treatment of refractory or unexplained chronic cough (RCC/UCC).[1] This condition, characterized by a persistent and debilitating cough, has long represented a significant unmet medical need with no specifically approved therapies in many parts of the world. Gefapixant introduces a novel therapeutic approach, moving beyond non-specific central suppression to the targeted modulation of peripheral neural pathways.

The drug's mechanism of action involves blocking adenosine triphosphate (ATP)-gated P2X3 ion channels located on sensory C fibers in the airway lining.[1] By interrupting this signaling pathway, which is believed to drive the neuronal hypersensitivity underlying chronic cough, Gefapixant aims to reduce the exaggerated cough reflex.[2] This targeted approach represents a significant evolution in the pharmacological management of cough.

The clinical development program was anchored by two large, pivotal Phase 3 trials, COUGH-1 and COUGH-2. In these studies, the 45 mg twice-daily (BID) dose of Gefapixant met its primary endpoint, demonstrating a statistically significant reduction in 24-hour cough frequency compared to placebo.[6] However, the therapeutic gain over a substantial placebo response was modest, a critical point that would later dominate regulatory discussions.[9]

The safety profile of Gefapixant is characterized by a high incidence of taste-related adverse events, including dysgeusia (altered taste), ageusia (loss of taste), and hypogeusia (reduced taste).[5] These on-target effects, while generally mild-to-moderate and reversible upon discontinuation, were frequent and led to higher treatment discontinuation rates compared to placebo.[7] The incidence of serious adverse events, however, was low and comparable to placebo.[7]

Perhaps the most compelling aspect of Gefapixant's journey is its divergent global regulatory outcomes. The drug has secured marketing authorization in major markets, including Japan (PMDA), Switzerland, and the European Union (EMA), where regulators determined that its benefits, although modest, outweighed its risks in a patient population with no approved alternatives.[2] In stark contrast, the U.S. Food and Drug Administration (FDA) repeatedly rejected the New Drug Application, citing a lack of substantial evidence of effectiveness and questioning the clinical meaningfulness of the observed efficacy.[14] This schism in regulatory assessment highlights a fundamental debate at the heart of modern drug development: the complex relationship between statistical significance and true clinical value, particularly for quality-of-life conditions.

II. Introduction: The Therapeutic Challenge of Chronic Cough and the Rationale for P2X3 Receptor Antagonism

Unmet Medical Need in Chronic Cough

Chronic cough, defined as a cough persisting for more than eight weeks, is a common and often debilitating condition affecting an estimated 5% to 10% of the adult population globally.[16] When a cough persists despite comprehensive investigation and appropriate treatment of underlying conditions such as asthma or gastroesophageal reflux disease, it is classified as refractory chronic cough (RCC). In cases where no underlying cause can be identified despite a thorough evaluation, it is termed unexplained chronic cough (UCC).[5] For millions of patients, RCC and UCC are not merely symptoms but chronic diseases that profoundly degrade quality of life, leading to physical exhaustion, social isolation, anxiety, depression, and even complications like stress urinary incontinence.[18]

Historically, the therapeutic landscape for these patients has been barren. In the absence of specifically approved treatments in many regions, clinicians have relied on off-label use of neuromodulators like gabapentin or opioids such as codeine and dextromethorphan.[2] While these agents can offer some relief, their use is often limited by significant side effects, including sedation, dizziness, and, in the case of opioids, the potential for abuse and dependence.[2] This has left a profound unmet medical need for a safe, effective, and targeted therapy for chronic cough.

The Cough Hypersensitivity Syndrome

The scientific understanding of chronic cough has evolved significantly, moving away from a purely reflexive model to the concept of a "cough hypersensitivity syndrome".[20] This paradigm posits that chronic cough is a clinical disorder rooted in neuronal dysfunction, characterized by an exaggerated, pathological cough reflex to low-level thermal, mechanical, or chemical stimuli that would not typically provoke a cough in healthy individuals.[21] This hypersensitization of the neural pathways governing the cough reflex provides a compelling scientific framework for the development of therapies that target these specific neuronal mechanisms rather than simply suppressing the motor act of coughing.

The Role of Purinergic Signaling

Research into the pathophysiology of cough hypersensitivity has identified purinergic signaling as a key pathway. In response to inflammation, irritation, or mechanical stress in the airways, epithelial cells release adenosine triphosphate (ATP) into the extracellular space.[4] This extracellular ATP functions as a crucial danger or damage signal, directly interacting with sensory nerve fibers that innervate the airway lining.[4]

P2X3 Receptors as a Therapeutic Target

The primary targets for this extracellular ATP on airway sensory nerves are P2X3 receptors.[4] P2X3 receptors are ligand-gated ion channels, predominantly expressed on the peripheral terminals of vagal afferent C fibers.[1] When ATP binds to these receptors, the ion channel opens, leading to cation influx, depolarization of the nerve fiber, and the generation of an action potential.[4] This signal is transmitted to the brainstem cough center, where it is perceived as an urge to cough, ultimately triggering the cough reflex motor pattern.[2] In chronic cough, it is hypothesized that excessive or prolonged activation of P2X3 receptors by ATP leads to the persistent neuronal hypersensitization that defines the condition.[21] This makes the P2X3 receptor a highly specific and rational target for therapeutic intervention.

Gefapixant's Rationale

Gefapixant was developed as a direct and logical intervention to interrupt this pathological signaling cascade. As a selective P2X3 receptor antagonist, it is designed to bind to these receptors and block their activation by extracellular ATP.[1] By preventing this initial step in the signaling pathway, Gefapixant aims to dampen the excessive activation of sensory C fibers, reduce the afferent nerve signals reaching the brain, and thereby normalize the hypersensitive cough reflex.[1] This mechanism represents a fundamental shift in antitussive therapy—a move from the non-specific central nervous system depression of traditional agents to a targeted, peripheral neuromodulation based on a modern, molecular understanding of the disease. It offers the promise of a novel, non-narcotic treatment that addresses a core pathophysiological driver of chronic cough.

III. Molecular Profile and Physicochemical Properties

Gefapixant is a small molecule drug belonging to the diaminopyrimidine class of compounds.[1] Its development and characterization have been extensively documented, providing a clear profile of its chemical and physical attributes.

Chemical Identity

The chemical structure of Gefapixant has been elucidated through various spectroscopic and crystallographic methods.[24] Its formal chemical name according to IUPAC nomenclature is 5-(2,4-diaminopyrimidin-5-yl)oxy-2-methoxy-4-propan-2-ylbenzenesulfonamide.[3] The molecule has a chemical formula of

C14​H19​N5​O4​S and a precise molecular weight of 353.4 g/mol.[3]

Physical Properties

In its solid state, Gefapixant presents as a white to pale yellow powder.[24] It has limited solubility in common laboratory solvents, being described as slightly soluble in dimethyl sulfoxide (DMSO).[26] Its calculated boiling point is 606.3±65.0 °C at 760 mmHg, and it has a density of approximately 1.4±0.1 g/cm³.[25]

Formulation

While the active moiety is the Gefapixant free base (CAS Number: 1015787-98-0), the drug substance used in the final commercial formulation is Gefapixant Citrate (CAS Number: 2310299-91-1).[21] This salt form was specifically selected during development to improve the drug's physicochemical properties and pharmacokinetic performance. In particular, the citrate salt was shown to enhance bioavailability and mitigate the effects of gastric pH changes, which can be caused by co-administration of medications such as proton pump inhibitors (PPIs).[24]

A consolidated summary of Gefapixant's key identifiers and properties is provided in Table 1.

Table 1: Gefapixant Identification and Chemical Properties

PropertyValueSource(s)
Non-proprietary NameGefapixant2
Brand NameLyfnua2
DrugBank IDDB150971
CAS Number1015787-98-0 (free base)3
ATC CodeR05DB291
SynonymsMK-7264, AF-219, RO-4926219, R-16462
Chemical FormulaC14​H19​N5​O4​S3
Molecular Weight353.4 g/mol21
IUPAC Name5-[(2,4-diaminopyrimidin-5-yl)oxy]-2-methoxy-4-(propan-2-yl)benzene-1-sulfonamide21
SMILES CodeO=S(C1=CC(OC2=CN=C(N)N=C2N)=C(C(C)C)C=C1OC)(N)=O21
InChI KeyHLWURFKMDLAKOD-UHFFFAOYSA-N21

IV. Preclinical and Clinical Pharmacology

The pharmacological profile of Gefapixant has been well-characterized through a combination of in vitro studies, animal models, and extensive clinical trials in humans. These studies have elucidated its precise mechanism of action, its effects on the cough reflex, and its behavior within the human body.

A. Mechanism of Action

Gefapixant's therapeutic effect is derived from its activity as a selective purinergic receptor antagonist.[1]

Primary Target and Selectivity

The primary molecular target of Gefapixant is the P2X3 purinoceptor, an ATP-gated ion channel.[1] It also demonstrates potent activity against the P2X2/3 heterotrimeric receptor, which is formed by a combination of P2X2 and P2X3 subunits.[23] The drug exhibits high potency, with half-maximal inhibitory concentration (

IC50​) values of approximately 30 nM against human P2X3 homotrimers and in the range of 100-250 nM at human P2X2/3 heterotrimeric receptors.[21] A key feature of its profile is its high selectivity; it has negligible activity (

IC50​ values >10,000 nM) at other P2X receptor subtypes, including P2X1, P2X2, P2X4, and P2X7, ensuring its action is focused on the intended target pathway.[25]

Allosteric Antagonism

Further mechanistic studies have revealed that Gefapixant functions as a reversible, negative allosteric modulator.[21] This is a crucial distinction from a competitive antagonist. Instead of competing with ATP for the same binding site, Gefapixant binds to a separate, allosteric site on the receptor protein. This binding induces a conformational change that reduces the channel's ability to open and conduct ions, even in the presence of high concentrations of the agonist (ATP).[21] This mode of action, which shows preferential activity at closed channels, allows it to effectively dampen receptor signaling independently of fluctuating local ATP levels.[23]

Physiological Effect

By binding to and inhibiting P2X3 and P2X2/3 receptors on the sensory C fibers of the vagus nerve in the airway, Gefapixant effectively blocks the ATP-mediated signaling that triggers the cough reflex.[1] This action is believed to reduce the underlying neuronal hypersensitivity, normalize the afferent signaling from the airways, and consequently decrease the frequency and urge to cough in patients with chronic cough.[4]

B. Pharmacodynamics

Clinical pharmacodynamic studies have provided direct evidence of Gefapixant's target engagement and its functional effects in humans.

Target Engagement

To confirm that Gefapixant acts on its intended target in a clinical setting, cough challenge studies were performed. In patients with chronic cough, treatment with Gefapixant was shown to significantly inhibit cough evoked by inhaled ATP.[22] One study reported a 4.7-fold increase in the concentration of ATP required to provoke a cough (C2 threshold) in patients treated with Gefapixant compared to placebo.[22] Critically, Gefapixant had no effect on cough induced by inhaled capsaicin or citric acid, which act through different sensory pathways (TRPV1 and acid-sensing ion channels, respectively).[22] This differential effect provides strong evidence of specific target engagement within the purinergic pathway in humans.

Clinical Onset and Duration

The therapeutic effects of Gefapixant are not immediate but build over time. Clinical trial data indicate that a significant reduction in 24-hour cough frequency compared to placebo becomes apparent by the fourth week of treatment and is sustained throughout the primary efficacy period of the studies.[1]

C. Pharmacokinetics (ADME)

The pharmacokinetic profile of Gefapixant—its absorption, distribution, metabolism, and excretion (ADME)—is favorable for a chronic oral medication, though it has one key characteristic that requires clinical attention.

Absorption

Following oral administration, Gefapixant is well-absorbed, with an estimated absolute bioavailability of at least 78%.[1] Peak plasma concentrations (

Tmax​) are typically reached between one and four hours post-dose.[1] At the recommended therapeutic dose of 45 mg twice daily, steady-state plasma concentrations are achieved within two days of initiating treatment.[1] Importantly for patient convenience, the co-administration of Gefapixant with a high-fat, high-calorie meal does not result in any clinically significant changes to its peak concentration (

Cmax​) or overall exposure (AUC), allowing it to be taken with or without food.[1]

Distribution

Gefapixant distributes into the tissues, with an estimated steady-state apparent volume of distribution (Vd​) of 133.8 L.[1] Its binding to plasma proteins is relatively low, at approximately 55%.[1] This low level of protein binding suggests that the risk of clinically significant drug-drug interactions resulting from displacement of other highly protein-bound drugs is minimal.

Metabolism

A significant advantage of Gefapixant's profile is that it undergoes minimal metabolism. The unchanged parent drug is the major drug-related component found in plasma, accounting for 87% of the total, with no single circulating metabolite accounting for more than 10%.[2] Overall, only about 14% of an administered dose is recovered as metabolites in the urine and feces.[2] The primary biotransformation pathways that do occur include hydroxylation, O-demethylation, dehydrogenation, oxidation, and direct glucuronidation.[2] Because metabolism is a minor route of elimination, the potential for drug-drug interactions involving cytochrome P450 enzymes is low.

Excretion

The primary route of elimination for Gefapixant and its metabolites is renal excretion.[1] Following a single oral radiolabeled dose, approximately 76.4% of the radioactivity was recovered in the urine, with another 22.6% recovered in the feces.[1] A substantial portion of the drug is excreted unchanged; parent drug accounted for 20% of the recovered dose in urine and 64% in feces.[1] The mean apparent terminal half-life (

t1/2​) ranges from 8.2 to 9.6 hours, a duration that supports a twice-daily dosing regimen.[29] The geometric mean apparent clearance (Cl/F) is approximately 10.8 L/h.[1]

This heavy reliance on renal excretion is the drug's primary pharmacokinetic liability. Any significant impairment in renal function can lead to drug accumulation and increased systemic exposure. This has been confirmed in population pharmacokinetic analyses and is the reason for the mandated dose adjustment in patients with severe renal impairment.[1] Thus, while its "clean" metabolic profile reduces the risk of many drug interactions, its dependence on the kidneys makes renal function the single most important patient factor to consider for safe prescribing.

A summary of key pharmacokinetic parameters is presented in Table 2.

Table 2: Summary of Pharmacokinetic Parameters of Gefapixant (45 mg BID)

ParameterValueSource(s)
Bioavailability (F)≥78%1
Time to Peak Concentration (Tmax​)1–4 hours1
Peak Concentration (Cmax​) at Steady-State531 ng/mL1
Area Under the Curve (AUC) at Steady-State4,144 ng∙hr/mL1
Volume of Distribution (Vd​)133.8 L1
Plasma Protein Binding~55%1
Apparent Terminal Half-life (t1/2​)8.2–9.6 hours29
Apparent Clearance (Cl/F)10.8 L/h1
Primary Route of EliminationRenal Excretion1

V. Clinical Efficacy in Refractory and Unexplained Chronic Cough

The efficacy of Gefapixant for the treatment of RCC and UCC was rigorously evaluated in a large-scale clinical development program, culminating in two pivotal Phase 3 trials. The results of these trials provided the core evidence for regulatory submissions worldwide and became the focal point of a significant scientific and regulatory debate.

A. The Phase 3 Program: Design of the COUGH-1 and COUGH-2 Trials

The cornerstones of the Gefapixant clinical program were two large, international, randomized, double-blind, placebo-controlled Phase 3 studies, designated COUGH-1 (NCT03449134) and COUGH-2 (NCT03449147).[6] These were landmark trials, representing the first companion Phase 3 studies ever conducted in the RCC/UCC patient population.[7]

Patient Population

The trials enrolled a total of 2,044 participants across 17-20 countries who had been diagnosed with RCC or UCC lasting for one year or longer.[9] The demographic profile of the participants closely mirrored that of patients seen in specialty cough clinics: predominantly female (~75%), with a mean age of 58 years, and a long history of suffering from the condition, with a mean cough duration of approximately 11 years.[7]

Treatment Arms and Duration

Participants were randomized in a 1:1:1 ratio to one of three treatment arms: placebo, Gefapixant 15 mg BID, or Gefapixant 45 mg BID.[6] The primary treatment periods, during which the main efficacy endpoints were assessed, differed slightly between the two studies: 12 weeks for COUGH-1 and 24 weeks for COUGH-2. These main periods were followed by blinded extension periods, bringing the total treatment duration up to 52 weeks for both trials.[7]

Endpoints

The primary efficacy endpoint in both trials was the change from baseline in 24-hour cough frequency, measured as coughs per hour.[7] This was measured objectively using the VitaloJAK™, an ambulatory digital audio recording device that captures sound recordings over a full 24-hour period for analysis.[6] Key secondary endpoints included awake cough frequency and several patient-reported outcomes (PROs) designed to capture the subjective burden of the disease, most notably the Leicester Cough Questionnaire (LCQ), a validated tool for assessing cough-specific quality of life.[11]

B. Efficacy in Reducing Cough Frequency

The results for the primary endpoint were consistent across both trials, demonstrating a statistically significant effect for the higher dose but also revealing the challenges of interpretation in this disease area.

Primary Endpoint Results

The Gefapixant 45 mg BID dose successfully met the primary endpoint in both COUGH-1 and COUGH-2, showing a statistically significant reduction in 24-hour cough frequency when compared to placebo.[7]

  • In COUGH-1, at the 12-week timepoint, treatment with Gefapixant 45 mg BID resulted in a placebo-adjusted relative reduction in cough frequency of 18.5% (95% CI: -32.9% to -0.9%; p=0.041).[8]
  • In COUGH-2, at the 24-week timepoint, the relative reduction versus placebo was 14.6% (95% CI: -26.1% to -1.4%; p=0.031).[8]

The Placebo Effect and the "Clinical Meaningfulness" Debate

While statistically significant, the magnitude of the treatment effect over placebo was modest. This was largely due to a robust and larger-than-anticipated placebo response observed in the trials.[9] For instance, while participants in the 45 mg group experienced an average reduction in cough frequency from baseline of 62-63%, those in the placebo group also saw a substantial reduction of 55-57%.[7]

This small therapeutic gain became the central issue during the U.S. FDA's review. The agency conducted its own post-hoc analyses, which calculated that the placebo-adjusted benefit amounted to an absolute reduction of only 1-2 coughs per hour.[33] This raised a critical question: is a statistically significant result of this magnitude also clinically meaningful for patients? The FDA ultimately concluded that it was not substantial enough to warrant approval, a decision that has had profound implications for the field.[10]

15 mg Dose Failure

In contrast to the higher dose, the Gefapixant 15 mg BID treatment arm failed to demonstrate a statistically significant reduction in cough frequency versus placebo in either of the Phase 3 trials.[9] This finding established 45 mg BID as the minimum effective dose.

C. Impact on Quality of Life and Patient-Reported Outcomes (PROs)

The trials also assessed the impact of treatment on patients' subjective experience of their condition.

Leicester Cough Questionnaire (LCQ) Results

Treatment with Gefapixant 45 mg BID led to improvements in patient-reported quality of life. A significantly greater proportion of participants in the 45 mg group achieved a clinically meaningful improvement in their LCQ total score (defined as an increase of ≥1.3 points) compared to those on placebo.[11] At the 24-week mark in a pooled analysis, 77.1% of patients receiving the 45 mg dose reported a clinically important improvement in their cough-related quality of life.[7] These improvements in PROs were shown to be maintained over the full 52-week treatment period.[11]

Correlation Issues

Despite these positive trends in PROs, the FDA's review highlighted a perceived lack of a clear correlation between the objective endpoint (the reduction in cough count) and the subjective endpoint (the improvement in patient well-being).[33] This disconnect further fueled regulatory skepticism, suggesting that the primary endpoint itself might not fully capture the aspects of the disease that are most burdensome to patients. This has led to calls within the research community to explore more nuanced endpoints for future chronic cough trials, such as the frequency and intensity of coughing bouts, which may better align with the patient experience.[33]

The Gefapixant clinical program, therefore, became an unintentional but landmark case study. While successful by traditional metrics of meeting pre-specified endpoints, it exposed the limitations of those very endpoints in a disease characterized by high placebo response and subjective burden, fundamentally altering the evidentiary standards for future therapies in this field.

A summary of the key efficacy findings is presented in Table 3.

Table 3: Key Efficacy Outcomes from COUGH-1 and COUGH-2 Trials (45 mg BID vs. Placebo)

OutcomeTrial (Timepoint)Gefapixant 45 mg BIDPlaceboPlacebo-Adjusted Relative Reduction (95% CI; p-value)Source(s)
24-Hour Cough FrequencyCOUGH-1 (Week 12)--18.5% (-32.9, -0.9; p=0.041)8
24-Hour Cough FrequencyCOUGH-2 (Week 24)--14.6% (-26.1, -1.4; p=0.031)8
% Reduction from Baseline (24-hr Cough Freq.)COUGH-1 (Week 12)62%55%-7
% Reduction from Baseline (24-hr Cough Freq.)COUGH-2 (Week 24)63%57%-7
% of Patients with Clinically Meaningful LCQ ImprovementPooled (Week 24)77.1%-Odds Ratio 1.41 (p=0.042) vs. Placebo7

VI. Safety and Tolerability Profile

The safety and tolerability of Gefapixant were extensively evaluated throughout its clinical development program, involving over 2,000 patients in the Phase 3 trials alone. The resulting profile is well-defined, characterized by a very common, dose-dependent, on-target adverse effect, but an otherwise favorable profile with respect to serious events.

A. Characterization of Adverse Events (AEs)

Most Common Adverse Events: Taste Disturbances

The most prominent and frequently reported adverse events associated with Gefapixant treatment are taste-related.[5] These encompass a spectrum of taste disturbances, formally categorized as:

  • Dysgeusia: An altered or distorted sense of taste.
  • Ageusia: A complete loss of the sense of taste.
  • Hypogeusia: A diminished or reduced sense of taste.[5]

The incidence of these events was highly dose-dependent. In the pivotal Phase 3 trials, the combined incidence of taste-related AEs in the 45 mg BID group was substantial, reported in 58% of patients in COUGH-1 and 69% in COUGH-2.[7] This was significantly higher than in the 15 mg BID group (~10-20%) and the placebo group (~3-8%).[7]

This side effect is not an idiosyncratic or off-target toxicity but is understood to be a direct, on-target pharmacological effect. Preclinical data have shown that P2X2 and P2X3 receptors are highly expressed on gustatory nerves and are essential for normal taste sensation.[11] Therefore, by antagonizing these receptors to reduce cough, Gefapixant concurrently antagonizes them in the taste buds, leading to the observed disturbances. This makes the side effect an inherent and inseparable part of the drug's mechanism of action, representing a fundamental trade-off between therapeutic benefit and tolerability.

Nature and Resolution of Taste Disturbances

The majority of these taste-related AEs were reported as mild to moderate in severity.[7] For most patients, the effect was transient. Pooled data from the long-term extensions of the Phase 3 trials showed that among participants who experienced a taste-related AE, the event had resolved in 96-99% of cases by the time of database lock.[11] Resolution typically occurred soon after treatment discontinuation, with a median time of 5 days.[34] Encouragingly for some patients, approximately 25% experienced resolution of the taste disturbance while continuing to take the drug.[11] However, it is noted in the European product information that in a few patients, these reactions persisted for more than a year after discontinuation.[34]

Other Common Adverse Events

Beyond taste disturbances, other adverse events reported more frequently than placebo included nausea and upper respiratory tract infections, although at a much lower incidence.[5]

B. Serious Adverse Events (SAEs), Discontinuations, and Special Precautions

Serious Adverse Events

The incidence of serious adverse events (SAEs) was low across the clinical program and, importantly, was similar between the Gefapixant treatment arms and the placebo group.[7] In the Phase 3 trials, the rate of SAEs was consistently below 4% in all groups.[7] The repeated rejections by the U.S. FDA were explicitly not related to safety concerns, underscoring the drug's favorable profile in this regard.[14]

Discontinuations due to Adverse Events

While not serious, the taste-related AEs were bothersome enough to impact treatment adherence. The rate of discontinuation due to adverse events was significantly higher in the 45 mg BID group, at 15% in COUGH-1 and 20% in COUGH-2.[7] This contrasts sharply with the discontinuation rates in the placebo groups, which were only 3% and 5%, respectively.[7] This highlights that the tolerability of the taste disturbance is a key factor in a patient's ability to remain on therapy long-term.

Special Precautions

The European Summary of Product Characteristics outlines several precautions for the use of Gefapixant:

  • Obstructive Sleep Apnea (OSA): A study using a supratherapeutic dose of Gefapixant (180 mg daily) in patients with moderate to severe OSA was associated with a lower mean oxygen saturation (SaO2​) during sleep. While the clinical relevance of this finding to the approved 45 mg BID dose is unknown, it is recommended that appropriate treatment for OSA be considered before initiating Gefapixant in patients with this comorbidity.[34]
  • Sulfonamide Moiety: Gefapixant's chemical structure contains a sulfonamide moiety. Although it is considered a non-sulphonylarylamine, it has not been studied in patients with a known history of severe hypersensitivity to sulfonamides. Therefore, cross-hypersensitivity cannot be definitively excluded, and the drug should be used with caution in this population.[34]

A summary of the incidence of key adverse events is presented in Table 4.

Table 4: Incidence of Common Adverse Events in COUGH-1 & COUGH-2

Adverse EventGefapixant 45 mg BID (%)Gefapixant 15 mg BID (%)Placebo (%)Source(s)
All Taste-Related AEs (Dysgeusia, Ageusia, Hypogeusia)58.0 – 68.610.7 – 19.53.3 – 8.37
NauseaCommonCommonCommon5
Upper Respiratory Tract InfectionCommonCommonCommon5
Discontinuations due to AEs15 – 203 – 83 – 57
Serious AEs< 4< 4< 47
Note: Nausea and URI are listed as common side effects, but specific percentages from Phase 3 trials were not detailed in the provided materials.

VII. Global Regulatory Landscape: A Tale of Divergent Assessments

The regulatory journey of Gefapixant is a compelling case study in the differing philosophies and evidentiary standards of the world's major health authorities. The same core data package, derived from the COUGH-1 and COUGH-2 trials, led to market authorization in Japan, Switzerland, and the European Union, but repeated rejection in the United States. This divergence underscores the complex and subjective nature of benefit-risk assessment for non-life-threatening, quality-of-life conditions.

A. Approval in Japan (PMDA) and Switzerland

Japan (PMDA)

Gefapixant, under the brand name Lyfnua, achieved its first global approval in Japan in January 2022 for the treatment of adults with refractory or unexplained chronic cough.[2] The decision followed a positive conclusion from the Second Committee on New Drugs on December 2, 2021.[24] The Pharmaceuticals and Medical Devices Agency (PMDA) review concluded that the product demonstrated efficacy and possessed an acceptable safety profile in view of its benefits.[24] The approval came with the condition of a post-marketing surveillance plan to further investigate the safety profile in real-world clinical practice, with a particular focus on the incidence of taste-related events.[24]

Switzerland (Swissmedic)

Shortly thereafter, Swissmedic, the Swiss agency for therapeutic products, granted marketing authorization for Lyfnua in May 2022.[2] The public summary report from Swissmedic noted that the studies showed a greater decrease in coughing frequency for patients treated with Lyfnua versus placebo and concluded that, taking all risks and precautions into account, the benefits of the drug outweighed the risks.[37]

These early approvals from two stringent regulatory bodies suggest a benefit-risk calculation that placed significant weight on the high unmet medical need in a therapeutic area with no approved treatments. They viewed the demonstrated efficacy, although modest, as sufficient and deemed the primary safety issue of taste disturbance to be a manageable risk for the target population.

B. Approval in the European Union (EMA)

The European Medicines Agency (EMA) followed a similar path. The Committee for Medicinal Products for Human Use (CHMP) issued a positive opinion recommending approval in July 2023.[31] This was followed by a full marketing authorization from the European Commission on September 15, 2023, making Lyfnua the first therapy to be approved for RCC or UCC in the European Union.[2]

The EMA's rationale, as detailed in its European Public Assessment Report (EPAR), is particularly illuminating. The agency explicitly acknowledged that "Lyfnua showed a modest effect in terms of reducing the number of daily coughs".[12] However, it concluded that the "benefits are greater than its risks" because the side effects were considered manageable and generally resolved after patients stopped treatment.[12] This statement encapsulates a regulatory philosophy that is willing to accept a modest benefit for a difficult-to-treat condition, provided the safety profile is acceptable, especially in the absence of any other approved therapeutic options.

C. Rejection in the United States (FDA)

In stark contrast, the U.S. Food and Drug Administration (FDA) reached the opposite conclusion on two separate occasions.

First Complete Response Letter (January 2022)

Following the initial New Drug Application (NDA) submission, the FDA issued a Complete Response Letter (CRL) in January 2022. A CRL indicates that the agency has completed its review but cannot approve the application in its present form. The FDA requested additional information on the efficacy of the drug.[10]

Resubmission and Advisory Committee Meeting (November 2023)

Merck conducted new analyses of the cough counting data and resubmitted the NDA. However, briefing documents released by the FDA ahead of a planned advisory committee meeting revealed that the agency remained skeptical about the clinical meaningfulness of the treatment effect.[10] In November 2023, the FDA's Pulmonary-Allergy Drugs Advisory Committee (PADAC) convened to discuss the application and voted overwhelmingly, 12-to-1, against recommending approval.[10] Panel members echoed the FDA's concerns, questioning whether the small treatment effect over placebo represented a significant enough difference for patients.[10]

Second Complete Response Letter (December 2023)

Following the negative advisory committee vote, the FDA issued a second and final CRL in December 2023.[14] The agency's formal conclusion was that the application "did not meet substantial evidence of effectiveness" for treating RCC and UCC.[14] The FDA clarified that the rejection was not based on safety concerns.[15] The core issues driving the FDA's decision were the small magnitude of the treatment effect on top of a large placebo response, the uncertainty about what constitutes a truly clinically meaningful benefit in chronic cough, and the perceived weak correlation between the objective cough count data and the subjective patient-reported outcomes.[10]

This tale of two continents demonstrates that regulatory approval is not merely a function of statistical significance but a complex judgment of value, context, and the interpretation of clinical meaningfulness, standards for which can clearly differ between even the most sophisticated global agencies.

A summary of the global regulatory status is provided in Table 5.

Table 5: Global Regulatory Status of Gefapixant

Regulatory AgencyRegionStatusDate of DecisionKey RationaleSource(s)
PMDAJapanApprovedJanuary 2022Efficacy demonstrated and safety acceptable in the context of high unmet need.13
SwissmedicSwitzerlandApprovedMay 2022Benefits of cough reduction outweigh the manageable risks (taste disturbance).36
EMAEuropean UnionApprovedSeptember 2023Acknowledged "modest effect" but deemed benefits greater than risks in a no-treatment area.2
FDAUnited StatesRejectedJan 2022 & Dec 2023Lack of substantial evidence of effectiveness; concerns over clinical meaningfulness of a small treatment effect.14

VIII. Dosing, Administration, and Clinical Application

For regions where Gefapixant (Lyfnua) is approved, the prescribing information provides clear guidelines for its use, including the standard dosing regimen and necessary adjustments for specific patient populations.

A. Recommended Dosing Regimen

Standard Dose

The recommended and approved dose of Gefapixant for adults with RCC or UCC is one 45 mg tablet taken orally twice daily.[34] This dose was established as the optimal balance of efficacy and tolerability based on the results of the Phase 2 and Phase 3 clinical trials, where the lower 15 mg BID dose was found to be ineffective.[9]

Administration

The tablets are intended to be swallowed whole and should not be broken, crushed, or chewed.[32] A key convenience for patients is that administration is not dependent on meals; the tablets may be taken with or without food.[35]

Missed Dose

Patients should be instructed that if a dose is missed, they should skip that dose entirely and take the next dose at its regularly scheduled time. They should not take two doses at once or otherwise attempt to make up for the missed dose.[34]

B. Dose Adjustments for Special Populations

The pharmacokinetic profile of Gefapixant, particularly its primary reliance on renal excretion, necessitates specific dose adjustments for certain patient populations to ensure safety and avoid excessive drug exposure.

Renal Impairment

Careful consideration of renal function is the most critical aspect of safe Gefapixant prescribing.

  • Severe Renal Impairment: For patients with severe renal impairment, defined as an estimated glomerular filtration rate (eGFR) of less than 30 mL/minute/1.73 m², who are not on dialysis, the dose must be reduced. The recommended dose in this population is one 45 mg tablet taken once daily.[34] Population pharmacokinetic simulations indicated that this once-daily regimen in patients with severe renal impairment results in a systemic exposure similar to that of the standard twice-daily regimen in patients with normal renal function.[28]
  • Mild to Moderate Renal Impairment: No dose adjustment is required for patients with mild or moderate renal impairment (eGFR ≥ 30 mL/minute/1.73 m²).[34]
  • End-Stage Renal Disease: There are insufficient data available for patients with end-stage renal disease requiring dialysis to make a formal dosing recommendation.[35]

Hepatic Impairment

Gefapixant has not been formally studied in patients with hepatic impairment. However, given that hepatic metabolism is a minor pathway for the drug's elimination, no dose adjustment is recommended for this population.[35]

Elderly (≥ 65 years)

No specific dose adjustment is required based on age alone.[34] However, clinicians should exercise caution, as elderly patients are more likely to have a natural decline in renal function. Because Gefapixant is substantially excreted by the kidneys, the risk of adverse reactions may be greater in older patients. Therefore, an assessment of renal function is particularly important when considering treatment in this age group.[34]

Pediatric Population

The safety and efficacy of Gefapixant have not been established in patients under 18 years of age. There is no relevant use for the drug in the pediatric population for the indication of RCC or UCC.[35]

IX. Synthesis and Future Outlook

The development, clinical evaluation, and regulatory odyssey of Gefapixant provide a multifaceted narrative with significant implications for the treatment of chronic cough and for pharmaceutical development more broadly. Its story is one of scientific innovation, clinical progress, and the evolving challenges of demonstrating value in a complex healthcare landscape.

A. Integrated Benefit-Risk Assessment

The core of the Gefapixant story lies in its benefit-risk profile, the interpretation of which has proven to be highly context-dependent.

Benefit

Gefapixant offers a clear and significant benefit as a first-in-class therapy with a novel, targeted, non-opioid mechanism of action for a debilitating condition that, in many parts of the world, has no approved treatments.[2] The clinical trial data robustly demonstrate that it provides a statistically significant reduction in cough frequency compared to placebo.[7] For a meaningful subset of patients, this translates into clinically important improvements in quality of life, which are sustained over the long term.[7] For individuals whose lives are severely disrupted by incessant coughing, even a modest reduction can represent a valuable therapeutic gain.

Risk

The primary risk associated with Gefapixant is the very high incidence of on-target taste disturbances.[5] While these adverse events are not medically serious and are reversible in the vast majority of patients, their high frequency and potential to be bothersome are significant enough to lead to treatment discontinuation in a substantial minority of patients (15-20%).[7] Other safety risks appear to be minimal, with rates of serious adverse events being low and comparable to placebo.[9]

Synthesis

The benefit-risk balance of Gefapixant is not absolute; it is a subjective judgment that hinges on the weight given to each component. The divergent global regulatory outcomes are a direct reflection of this subjectivity. For regulatory agencies like the EMA and PMDA, the benefit of providing the first approved therapy for a long-neglected condition, even with modest efficacy, outweighed the manageable risk of a non-serious but common side effect.[12] For a regulator like the FDA, which holds a high bar for "substantial evidence of effectiveness," the modest therapeutic gain over placebo was deemed insufficient to justify approval, regardless of the manageable safety profile.[10] Ultimately, for the individual patient, the decision to use Gefapixant (where available) will involve a personal calculation: whether the potential for cough relief is worth the high probability of experiencing altered taste.

B. The Future of P2X3 Antagonism and Chronic Cough Therapy

Regardless of its regulatory fate in the U.S., the development of Gefapixant has irrevocably changed the landscape of chronic cough therapy.

Pioneering a Class

Gefapixant has successfully validated the P2X3 receptor as a legitimate and druggable therapeutic target for chronic cough. Its journey from bench to bedside has provided the proof-of-concept for an entire new class of medicines. This has paved the way for competitors, such as GSK's camlipixant and other P2X3 antagonists in development, which may seek to offer an improved balance of efficacy and tolerability.[15]

Lessons for Clinical Development

The regulatory challenges faced by Gefapixant offer critical lessons for the entire field. Its story demonstrates that in modern drug development, particularly for quality-of-life conditions, achieving statistical significance on a traditional endpoint is no longer a guaranteed path to approval. Future clinical development programs for chronic cough and similar conditions will likely need to evolve in several key ways:

  1. Endpoint Innovation: There will be a strong impetus to move beyond simple 24-hour cough frequency. Future trials may need to incorporate and validate more nuanced endpoints, such as the analysis of cough bouts, cough intensity, and other measures that might correlate more closely with patient-reported burden and quality of life.[33]
  2. Demonstrating Clinical Meaningfulness: Programs will need to prospectively design studies to rigorously demonstrate that the observed treatment effect is not just statistically significant but also clinically meaningful from the patient's perspective, a standard that the FDA felt Gefapixant did not meet.
  3. Managing the Placebo Effect: A deeper understanding and better strategies for managing the high placebo response inherent in this condition will be crucial for designing trials that can show a clearer and more robust treatment effect.

Place in Therapy

In the European Union, Japan, and other regions where it is approved, Gefapixant is positioned to become an important new tool for physicians treating patients with RCC and UCC. It will likely be used as a second- or third-line agent for patients who have failed to respond to the investigation and treatment of underlying conditions. Its ultimate success in the marketplace will depend on real-world effectiveness, patient persistence in the face of taste-related side effects, and the eventual arrival of next-generation P2X3 antagonists.

In conclusion, the story of Gefapixant is a microcosm of the evolving challenges and complexities of pharmaceutical innovation. It highlights a shift where demonstrating biological activity and achieving statistical significance are necessary but no longer sufficient conditions for success. The new frontier is the ability to prove a drug's comprehensive value proposition to a diverse array of stakeholders—regulators, payers, clinicians, and patients—each with their own definition of what constitutes a meaningful benefit. The lessons learned from Gefapixant's journey will undoubtedly shape the future of drug development for chronic cough and beyond.

Works cited

  1. Gefapixant | C14H19N5O4S | CID 24764487 - PubChem, accessed August 26, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/24764487
  2. Gefapixant: Uses, Interactions, Mechanism of Action | DrugBank ..., accessed August 26, 2025, https://go.drugbank.com/drugs/DB15097
  3. Gefapixant - Wikipedia, accessed August 26, 2025, https://en.wikipedia.org/wiki/Gefapixant
  4. gefapixant (Pending FDA Approval) - Medscape Reference, accessed August 26, 2025, https://reference.medscape.com/drug/gefapixant-4000201
  5. P2X3 antagonist: Drug Class, Uses, Side Effects, Drug Names - RxList, accessed August 26, 2025, https://www.rxlist.com/p2x3_antagonist/drug-class.htm
  6. A Study Looks at Gefapixant Treatment for Chronic Cough | Respiratory Therapy, accessed August 26, 2025, https://respiratory-therapy.com/products-treatment/gefapixant-treatment-chronic-cough/
  7. Merck's Gefapixant (45 mg Twice Daily) Significantly Decreased Cough Frequency Compared to Placebo at Week 12 and 24 in Patients with Refractory or Unexplained Chronic Cough, accessed August 26, 2025, https://www.merck.com/news/mercks-gefapixant-45-mg-twice-daily-significantly-decreased-cough-frequency-compared-to-placebo-at-week-12-and-24-in-patients-with-refractory-or-unexplained-chronic-cough/
  8. Efficacy and safety of gefapixant, a P2X3 receptor antagonist, in ..., accessed August 26, 2025, https://pureadmin.qub.ac.uk/ws/files/301883845/Cough1_and_Cough2_5_3_21_Resubmission_Clean_8_13_.pdf
  9. Late Breaking Abstract - Two Phase 3 Randomized Clinical Trials of Gefapixant, a P2X3 Receptor Antagonist, in Refractory or Unexplained Chronic Cough (COUGH-1 and COUGH-2) - ERS Publications, accessed August 26, 2025, https://publications.ersnet.org/content/erj/56/suppl64/3800
  10. UPDATE: FDA AdComm shoots down Merck's chronic cough med in 12-1 vote - Fierce Biotech, accessed August 26, 2025, https://www.fiercebiotech.com/biotech/fda-adcomm-shoots-down-mercks-chronic-cough-med-12-1-vote
  11. Efficacy and Safety of Gefapixant for Refractory or Unexplained Chronic Cough over 52 Weeks - ATS Journals, accessed August 26, 2025, https://www.atsjournals.org/doi/10.1164/rccm.202211-2128LE
  12. Lyfnua | European Medicines Agency (EMA), accessed August 26, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/lyfnua
  13. Gefapixant: First Approval - PubMed, accessed August 26, 2025, https://pubmed.ncbi.nlm.nih.gov/35347635/
  14. Merck Provides U.S. Regulatory Update on Gefapixant - Merck.com, accessed August 26, 2025, https://www.merck.com/news/merck-provides-u-s-regulatory-update-on-gefapixant/
  15. Merck's Chronic Cough Drug Fails to Secure FDA Approval for Second Time - BioSpace, accessed August 26, 2025, https://www.biospace.com/merck-s-chronic-cough-drug-fails-to-secure-fda-approval-for-second-time
  16. FDA rejects MSD's gefapixant for chronic cough - Pharmaceutical Technology, accessed August 26, 2025, https://www.pharmaceutical-technology.com/news/fda-rejects-msds-gefapixant-for-chronic-cough/
  17. Merck's Gefapixant Drug For Chronic Cough A Step Away From Approval In Europe, accessed August 26, 2025, https://www.nasdaq.com/articles/mercks-gefapixant-drug-for-chronic-cough-a-step-away-from-approval-in-europe
  18. Gefapixant Completed Phase 3 Trials for Chronic Cough (CC) Treatment | DrugBank Online, accessed August 26, 2025, https://go.drugbank.com/drugs/DB15097/clinical_trials?conditions=DBCOND0085805&phase=3&purpose=treatment&status=completed
  19. Exploring the Potential of a P2X3 Receptor Antagonist: Gefapixant in the Management of Persistent Cough Associated with Interstitial Lung Disease - MDPI, accessed August 26, 2025, https://www.mdpi.com/1648-9144/61/5/892
  20. Efficacy and safety of gefapixant for chronic cough: a meta-analysis of randomised controlled trials - ERS Publications, accessed August 26, 2025, https://publications.ersnet.org/content/errev/32/168/220219
  21. Gefapixant | AF-219 | CAS#1015787-98-0 | P2X3 antagonist - MedKoo Biosciences, accessed August 26, 2025, https://www.medkoo.com/products/17250
  22. The Effect of Gefapixant, a P2X3 antagonist, on Cough Reflex Sensitivity: A randomised placebo-controlled study - Hull Repository, accessed August 26, 2025, https://hull-repository.worktribe.com/file/1835195/1/Article
  23. GEFAPIXANT (PD057987, HLWURFKMDLAKOD-UHFFFAOYSA-N) - Probes & Drugs, accessed August 26, 2025, https://www.probes-drugs.org/compound/PD057987/
  24. Report on the Deliberation Results December 8, 2021 ..., accessed August 26, 2025, https://www.pmda.go.jp/files/000263285.pdf
  25. CAS 1015787-98-0 Gefapixant - BOC Sciences, accessed August 26, 2025, https://www.bocsci.com/product/gefapixant-cas-1015787-98-0-138428.html
  26. Gefapixant (AF-219, MK-7264, Ro 4926219, CAS Number: 1015787-98-0), accessed August 26, 2025, https://www.caymanchem.com/product/40825/gefapixant
  27. Gefapixant (MK-7264) | P2X3 Antagonist - MedchemExpress.com, accessed August 26, 2025, https://www.medchemexpress.com/Gefapixant.html
  28. Proposed Structures of Human Metabolites of gefapixant | Download ..., accessed August 26, 2025, https://www.researchgate.net/figure/Proposed-Structures-of-Human-Metabolites-of-gefapixant_fig3_358278624
  29. (PDF) Single‐ and Multiple‐Dose Pharmacokinetics of Gefapixant (MK‐7264), a P2X3 Receptor Antagonist, in Healthy Adults - ResearchGate, accessed August 26, 2025, https://www.researchgate.net/publication/380033586_Single-_and_Multiple-Dose_Pharmacokinetics_of_Gefapixant_MK-7264_a_P2X3_Receptor_Antagonist_in_Healthy_Adults
  30. Single- and Multiple-Dose Pharmacokinetics of Gefapixant (MK-7264), a P2X3 Receptor Antagonist, in Healthy Adults - PubMed, accessed August 26, 2025, https://pubmed.ncbi.nlm.nih.gov/38651193/
  31. Gefapixant Gets European Medicines Agency's Backing for Chronic Cough, accessed August 26, 2025, https://respiratory-therapy.com/disorders-diseases/chronic-pulmonary-disorders/chronic-diseases/gefapixant-european-medicines-agencys-backing-chronic-cough/
  32. Public Assessment Report National Procedure Lyfnua 45 mg film-coated tablets gefapixant citrate PLGB 53095/0091 MERCK SHARP &am - NET, accessed August 26, 2025, https://mhraproducts4853.blob.core.windows.net/docs/b7d1d94a2ef609c09b1b0c48de1811fef5ea74eb
  33. Unpacking the FDA Advisory Committee Meeting for Merck's Gefapixant - Strados Labs, accessed August 26, 2025, https://stradoslabs.com/unpacking-the-fda-advisory-committee-meeting-for-mercks-gefapixant/
  34. LYFNUA, INN-gefapixant citrate - European Medicines Agency, accessed August 26, 2025, https://www.ema.europa.eu/en/documents/product-information/lyfnua-epar-product-information_en.pdf
  35. Lyfnua 45 mg film-coated tablets - Summary of Product Characteristics (SmPC) - (emc), accessed August 26, 2025, https://www.medicines.org.uk/emc/product/15778/smpc
  36. SwissPAR - Lyfnua - Swissmedic, accessed August 26, 2025, https://www.swissmedic.ch/dam/swissmedic/de/dokumente/zulassung/swisspar/68065-lyfnua-01-swisspar-20220812.pdf.download.pdf/SwissPAR_Lyfnua.pdf
  37. Public Summary SwissPAR – Lyfnua® - Swissmedic, accessed August 26, 2025, https://www.swissmedic.ch/swissmedic/en/home/about-us/publications/public-summary-swiss-par/public-summary-swiss-par-lyfnua.html
  38. Merck Receives Positive European Union CHMP Opinion for Gefapixant, accessed August 26, 2025, https://www.americanpharmaceuticalreview.com/1315-News/598632-Merck-Receives-Positive-European-Union-CHMP-Opinion-for-Gefapixant/
  39. Fona DS Common - Square Pharmaceuticals, accessed August 26, 2025, https://www.squarepharma.com.bd/downloads/1728297621_pdoc_Ds%20Gefapix%20for%20Website.pdf

Published at: August 26, 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.