MedPath

Vonoprazan Advanced Drug Monograph

Published:Aug 3, 2025

Generic Name

Vonoprazan

Brand Names

Voquezna 14 Day Dualpak 20;500, Voquezna 14 Day Triplepak 20;500;500

Drug Type

Small Molecule

Chemical Formula

C17H16FN3O2S

CAS Number

881681-00-1

Associated Conditions

Duodenal Ulcer, Gastric Ulcer, Gastric or Duodenal Ulcers Caused by Low-dose Aspirin, Helicobacter Pylori Infection, Reflux Esophagitis (RE), Develop NSAID-induced gastric ulcers

Comprehensive Report on Vonoprazan: A First-in-Class Potassium-Competitive Acid Blocker

I. Introduction: A Paradigm Shift in Acid Suppression Therapy

The global burden of acid-related gastrointestinal disorders, including gastroesophageal reflux disease (GERD), peptic ulcer disease (PUD), and infections caused by the bacterium Helicobacter pylori, represents a significant and persistent challenge to public health and clinical practice.[1] For decades, the therapeutic landscape for these conditions has been dominated by proton pump inhibitors (PPIs), a class of drugs that includes well-known agents such as omeprazole and lansoprazole. The advent of PPIs marked a major advance in gastroenterology, offering effective acid suppression that became the standard of care worldwide.[4]

However, despite their widespread success, the pharmacological profile of PPIs is characterized by several well-documented limitations that can compromise their clinical utility. A fundamental aspect of their mechanism is the requirement for activation by acid within the parietal cell canaliculi. This necessitates that PPIs, which are unstable in acidic environments and require enteric coating, be administered 30 to 60 minutes before a meal to ensure they are present in the bloodstream when the proton pumps are maximally stimulated by food intake.[4] Furthermore, PPIs often require several days of repeated dosing to achieve their full therapeutic effect, a delay that can be suboptimal for patients seeking rapid symptom relief.[1] Perhaps one of the most significant clinical challenges is their inability to provide complete 24-hour acid control, with many patients experiencing nocturnal acid breakthrough, a phenomenon where gastric pH drops during the night despite appropriate dosing.[6] This incomplete suppression is linked to the irreversible nature of their binding to active proton pumps and the daily turnover of new, uninhibited pumps.

Compounding these kinetic limitations is the issue of inter-individual variability. PPIs are primarily metabolized by the cytochrome P450 enzyme CYP2C19, which is subject to significant genetic polymorphism. A patient's genotype can categorize them as a poor, intermediate, normal, rapid, or ultra-rapid metabolizer, leading to highly variable drug exposure and clinical response. This can result in therapeutic failure in rapid metabolizers and an increased risk of adverse effects in poor metabolizers, creating unpredictability in clinical outcomes.[5]

In response to these unmet needs, a new class of drugs, the potassium-competitive acid blockers (P-CABs), was developed. These agents were engineered to overcome the inherent pharmacological weaknesses of PPIs.[1] Vonoprazan stands as a first-in-class P-CAB in many global markets, representing not merely an incremental improvement but a fundamental paradigm shift in the management of acid-related disorders.[2] The development and successful commercialization of vonoprazan reflect a deliberate and strategic effort to address the long-standing clinical frustrations associated with PPI therapy. By designing a molecule with a distinct mechanism of action—one that is acid-stable, acts rapidly, provides sustained inhibition, and is independent of CYP2C19 metabolizer status—its developers targeted the specific vulnerabilities of the previous generation of acid suppressants. The strong market performance of vonoprazan, particularly in Japan where it generated over $500 million in net sales in its fourth full year, validates the clinical and commercial value of this innovative approach, demonstrating that a deep understanding of existing therapeutic gaps can drive significant advances in patient care.[19]

II. Vonoprazan: Chemical Profile and Physicochemical Characteristics

Vonoprazan is a synthetic, small molecule drug classified chemically as a pyrrole derivative.[10] Its unique structure is central to its novel mechanism of action and distinguishes it from the benzimidazole structure of traditional PPIs.

Chemical Identity

The systematic International Union of Pure and Applied Chemistry (IUPAC) name for vonoprazan is 1-[5-(2-Fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine.[21] This name precisely describes its molecular architecture, which features a central pyrrole ring substituted with a fluorophenyl group, a pyridinylsulfonyl group, and a methylaminomethyl side chain.

Identifiers and Formulations

For scientific and regulatory purposes, vonoprazan is identified by several unique codes. The Chemical Abstracts Service (CAS) has assigned the number 881681-00-1 to the vonoprazan free base. However, the drug is formulated for clinical use as a fumarate salt, which has the distinct CAS Number 1260141-27-2.[10] This distinction is critical when reviewing scientific literature and regulatory documents.

The molecular formula for the free base is C17​H16​FN3​O2​S, corresponding to a molecular weight of approximately 345.4 g/mol.[10] The fumarate salt, which incorporates one molecule of fumaric acid (

C4​H4​O4​), has a combined molecular formula of C17​H16​FN3​O2​S⋅C4​H4​O4​ (also written as C21​H20​FN3​O6​S) and a molecular weight of 461.47 g/mol.[15]

Key identifiers in major pharmacological and chemical databases include:

  • DrugBank ID: DB11739 [22]
  • PubChem CID: 15981397 [22]
  • ChEMBL ID: CHEMBL2079130 (free base) [22]

Physical and Chemical Properties

Vonoprazan is a synthetic compound that exists as a white to off-white solid powder.[10] Its solubility profile is characterized by being very slightly soluble in water and methanol, but soluble in dimethyl sulfoxide (DMSO).[10] For laboratory and long-term storage, it is recommended to be kept in a freezer at -20°C, protected from light in an inert atmosphere.[10]

A pivotal physicochemical property of vonoprazan is its high basicity, reflected in its pKa value. Reported values are consistently high, ranging from 8.94 to 9.06.[15] This high pKa means that the molecule is readily protonated (becomes positively charged) in acidic environments, a feature that is fundamental to its potent pharmacodynamic effect and its mechanism of accumulation in gastric parietal cells.

The following table provides a consolidated summary of the key identification and physicochemical properties of vonoprazan, clarifying the distinction between the free base and the clinically relevant fumarate salt.

Table 1: Summary of Physicochemical and Identification Properties of Vonoprazan

PropertyVonoprazan (Free Base)Vonoprazan FumarateSource(s)
IUPAC Name1-[5-(2-Fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamineNot Applicable21
CAS Number881681-00-11260141-27-210
Molecular FormulaC17​H16​FN3​O2​SC17​H16​FN3​O2​S⋅C4​H4​O4​15
Molecular Weight345.40 g/mol461.47 g/mol15
AppearanceWhite to off-white powder/solidWhite to off-white powder15
SolubilitySoluble in DMSOVery slightly soluble in water and methanol10
pKa8.94 - 9.069.0615

III. Pharmacological Profile: Mechanism of Potent and Sustained Acid Inhibition

Vonoprazan's therapeutic efficacy stems from its distinct pharmacological profile as a potassium-competitive acid blocker (P-CAB), a classification that sets it apart from traditional PPIs.[1] Its mechanism of action is characterized by direct, potent, and sustained inhibition of the final step in the gastric acid secretion pathway.

Target and Binding Mechanism

The molecular target of vonoprazan is the gastric hydrogen-potassium adenosine triphosphatase (H+/K+-ATPase) enzyme system, commonly referred to as the proton pump. This enzyme is embedded in the membrane of the secretory canaliculi of gastric parietal cells and is responsible for pumping hydrogen ions (H+) into the gastric lumen in exchange for potassium ions (K+).[6]

Vonoprazan functions through a mechanism of competitive and reversible inhibition. It binds ionically to the K+-binding site on the luminal side of the proton pump, directly and competitively obstructing the access of potassium ions.[7] This blockade prevents the conformational change in the enzyme that is necessary for the transport of

H+ ions, thereby inhibiting acid secretion.[6] This mode of action contrasts sharply with that of PPIs, which require acid-catalyzed conversion to a reactive sulfenamide intermediate that then forms an irreversible, covalent disulfide bond with cysteine residues on the enzyme.[7]

Pharmacodynamic Advantages over PPIs

The unique chemical properties of vonoprazan confer several significant pharmacodynamic advantages that directly address the limitations of PPIs.

  • Acid Stability and Rapid Onset of Action: Vonoprazan is a stable base that does not require an acidic environment for its activation. Unlike PPIs, which are acid-labile and must be protected by enteric coatings until they reach the small intestine for absorption, vonoprazan can exert its effect directly upon reaching the parietal cell.[6] Because its binding is not dependent on the pump being in an active, acid-secreting state, it can inhibit both active and resting proton pumps. This results in a remarkably rapid onset of action, with significant acid suppression achieved within hours of the first dose, a stark contrast to the several days required for PPIs to reach maximal effect.[1]
  • High Accumulation and Prolonged Duration of Action: The high pKa of vonoprazan (9.06) is a key determinant of its potency and duration. When vonoprazan, a weak base, enters the highly acidic environment of the parietal cell secretory canaliculi (where pH can be less than 2), it rapidly becomes protonated, acquiring a positive charge.[7] This charged form is unable to readily diffuse back across the cell membrane, effectively "trapping" the drug at its site of action. This leads to a massive accumulation of vonoprazan in the canaliculi, with concentrations estimated to be over 100,000-fold higher than in the plasma.[7] This high local concentration, combined with a slow clearance rate from the gastric glands and a relatively long plasma half-life, ensures a potent and sustained acid-suppressive effect that persists for over 24 hours following a single dose.[6]
  • High Potency: In vitro studies have quantified the superior potency of vonoprazan. It inhibits H+/K+-ATPase with a half-maximal inhibitory concentration (IC50​) of 19 nM at a pH of 6.5.[10] In direct comparisons, its inhibitory potency has been shown to be approximately 350 times higher than that of the PPI lansoprazole.[12]

The pharmacology of vonoprazan can be seen as an elegant solution to the kinetic challenges posed by PPIs. The combination of a high pKa and reversible binding is particularly noteworthy. While PPIs rely on irreversible binding, their effect diminishes as new proton pumps are synthesized throughout the day, contributing to phenomena like nocturnal acid breakthrough.[4] Vonoprazan's mechanism circumvents this limitation. The high pKa ensures that a concentrated depot of the drug is maintained at the site of action. The reversible nature of its binding allows molecules that dissociate from one pump to remain within this highly concentrated local environment, ready to rapidly engage and inhibit other pumps, including those newly synthesized. This dynamic process of "trapping and re-engagement" creates a more stable and continuous state of inhibition over a 24-hour period, one that is less dependent on pump activation status or precise mealtime dosing. This sophisticated mechanism is the foundation of vonoprazan's ability to provide more rapid, potent, and durable acid control than its predecessors.

IV. Comprehensive Pharmacokinetic (PK) Profile

The pharmacokinetic profile of vonoprazan underpins its clinical advantages, particularly its predictable behavior and dosing convenience. Its absorption, distribution, metabolism, and excretion (ADME) properties have been well-characterized in numerous clinical studies.

Absorption

Following oral administration, vonoprazan is rapidly absorbed from the gastrointestinal tract. The time to reach maximum plasma concentration (Tmax) is typically observed between 1.5 and 3.0 hours.[7] A key clinical advantage is the minimal impact of food on its absorption. While a high-fat meal was observed to slightly increase the maximum plasma concentration (Cmax) by 5% and the total exposure (Area Under the Curve, AUC) by 15%, these changes are not considered clinically significant.[7] This allows vonoprazan to be administered without regard to mealtimes, a major departure from the strict pre-meal dosing required for most PPIs and a significant improvement in patient convenience and adherence.[7]

Distribution

Vonoprazan exhibits extensive distribution into tissues, which is reflected by its large apparent volume of distribution (Vd) of approximately 783 to 1050 L.[7] It is highly bound to plasma proteins, with binding ranging from 85% to 88% in healthy subjects; this binding is not saturated at therapeutic concentrations.[7] As described in its pharmacodynamic profile, the most pharmacologically significant aspect of its distribution is its high degree of accumulation and concentration within the acidic canaliculi of gastric parietal cells, its site of action.[7]

Metabolism

Vonoprazan undergoes extensive metabolism, resulting in the formation of pharmacologically inactive metabolites.[8] The metabolism involves multiple pathways, which reduces its reliance on any single enzyme.

  • Primary Pathway: The main metabolic route is oxidation via the cytochrome P450 (CYP) 3A4 enzyme.[8]
  • Minor Pathways: To a lesser extent, vonoprazan is also metabolized by several other CYP isoforms, including CYP2B6, CYP2C19, CYP2D6, and CYP2C9.[8]
  • Non-CYP Pathway: In addition to CYP-mediated oxidation, a non-oxidative pathway involving sulfotransferase SULT2A1 also contributes to its metabolism.[12]

Excretion

The elimination of vonoprazan is characterized by a mean apparent terminal half-life (t1/2) of approximately 7 to 9 hours.[7] This is substantially longer than the 1- to 2-hour half-life of most PPIs and is a key contributor to its prolonged duration of action. The drug reaches steady-state plasma concentrations after 3 to 4 days of once-daily dosing.[7]

Excretion of the metabolites occurs primarily through the kidneys, with approximately 67% of an administered dose recovered in the urine. The remaining 31% is excreted in the feces.[7] Only a small fraction (around 8%) of the drug is excreted unchanged in the urine, underscoring the extensive nature of its metabolism.[12]

Population Pharmacokinetics (PopPK)

A comprehensive population pharmacokinetic analysis, integrating data from 15 clinical trials involving 746 patients and 410 healthy volunteers, has provided robust insights into factors that might influence drug exposure.[25] This analysis confirmed that while covariates such as race (Asian vs. non-Asian), sex, age, body weight, and disease status had a statistically identifiable effect on vonoprazan's pharmacokinetics, the magnitude of these effects was small and not considered clinically relevant. Consequently, no dose adjustments are recommended based on these demographic or patient characteristics, highlighting the drug's predictable behavior across diverse populations.[25]

The following table summarizes the key pharmacokinetic parameters of vonoprazan, providing a concise reference for its ADME profile.

Table 2: Summary of Key Pharmacokinetic Parameters of Vonoprazan

ParameterValueSource(s)
Time to Max. Concentration (Tmax)1.5 - 3.0 hours7
Max. Concentration (Cmax, 20 mg dose)~23.3 - 37.8 ng/mL7
Area Under Curve (AUC0-12h, 20 mg BID)~273 ng·hr/mL7
Elimination Half-Life (t1/2)~7 - 9 hours7
Volume of Distribution (Vd)783 - 1050 L7
Plasma Protein Binding85% - 88%7
Primary Metabolic PathwayCYP3A48
Excretion Routes~67% Urine (as metabolites), ~31% Feces7
Food EffectNot clinically significant7

V. Clinical Evidence and Therapeutic Efficacy

The clinical development program for vonoprazan has been extensive, with numerous Phase 3 trials establishing its efficacy and safety across a range of acid-related disorders. It has consistently demonstrated non-inferiority and, in several key endpoints, superiority to standard-of-care PPIs.

A. Healing and Maintenance of Erosive Esophagitis (EE) and GERD

  • Healing of Erosive Esophagitis: The efficacy of vonoprazan in healing EE has been robustly demonstrated. The pivotal Phase 3 PHALCON-EE trial (NCT04124926), a large, randomized, double-blind study, compared vonoprazan 20 mg once daily to lansoprazole 30 mg once daily in over 1000 patients with all grades of EE.[7] Vonoprazan met the primary endpoint of non-inferiority for the rate of complete endoscopic healing by Week 8. Furthermore, in a pre-specified secondary analysis, vonoprazan was found to be superior to lansoprazole, with a healing rate of 92.9% versus 84.6%, respectively.[7] The superiority was particularly pronounced in patients with more severe disease (Los Angeles Classification Grades C and D), an area of significant unmet need with PPI therapy.[9]
  • Maintenance of Healed Erosive Esophagitis: For patients requiring long-term therapy to prevent recurrence, vonoprazan has also proven superior to PPIs. In the maintenance phase of the PHALCON-EE trial, patients who achieved healing were re-randomized to receive either vonoprazan 10 mg, vonoprazan 20 mg, or lansoprazole 15 mg once daily for 24 weeks.[7] Vonoprazan 10 mg was superior to lansoprazole 15 mg in maintaining endoscopic healing at 6 months (79.2% vs. 72.0%), providing a more durable remission for patients.[7]
  • Non-Erosive Reflux Disease (NERD): Vonoprazan 10 mg daily is also approved for the treatment of heartburn associated with NERD, the most common GERD phenotype, expanding its utility to a broader patient population.[34]
  • PPI-Refractory GERD: A growing body of evidence supports the use of vonoprazan in patients who have an inadequate response to standard or high-dose PPI therapy. Its potent and sustained acid suppression mechanism makes it an effective option for this challenging-to-treat patient group.[6]

B. Eradication of Helicobacter pylori Infection

Vonoprazan has become a cornerstone of modern H. pylori eradication strategies, demonstrating higher efficacy rates than PPI-based regimens, particularly in the face of rising antibiotic resistance. It is available in the U.S. as co-packaged products, Voquezna Triple Pak and Voquezna Dual Pak, to improve convenience and adherence.[35]

  • Vonoprazan-Based Triple Therapy: The standard triple therapy consists of vonoprazan 20 mg twice daily, amoxicillin 1000 mg twice daily, and clarithromycin 500 mg twice daily for 14 days. In the Phase 3 PHALCON-HP trial, this regimen demonstrated a higher eradication rate than the lansoprazole-based triple therapy (84.7% vs. 78.8%) in patients without clarithromycin or amoxicillin resistance.[7] Crucially, in patients with known clarithromycin-resistant H. pylori strains, the vonoprazan-based regimen was significantly more effective (65.8% vs. 31.9% for the lansoprazole regimen), addressing a major cause of treatment failure globally.[4] Multiple studies have confirmed eradication rates exceeding 90% with vonoprazan-based triple therapy.[4]
  • Vonoprazan-Based Dual Therapy: A novel and important regimen is the dual therapy, comprising vonoprazan 20 mg twice daily and amoxicillin 1000 mg three times daily for 14 days. This clarithromycin-sparing regimen was shown to be non-inferior to lansoprazole triple therapy in the PHALCON-HP trial and offers a valuable alternative for patients with clarithromycin allergy or in regions with high resistance rates.[7]

C. Prevention of NSAID and Low-Dose Aspirin-Induced Ulcers

In Japan, where vonoprazan has been in use the longest, it is approved and widely used for the prevention of recurrent gastric or duodenal ulcers in patients requiring long-term therapy with non-steroidal anti-inflammatory drugs (NSAIDs) or low-dose aspirin.[29] Clinical trials in Japanese patients have demonstrated that vonoprazan 10 mg daily is effective for this indication, providing crucial gastroprotection for at-risk populations.[12]

The table below summarizes key clinical trials that form the evidentiary basis for vonoprazan's approved indications.

Table 3: Summary of Pivotal Phase 3 Clinical Trials for Vonoprazan

Trial Identifier / Lead AuthorIndicationPhaseDesignArmsPrimary EndpointKey Results / ConclusionSource(s)
PHALCON-EE (NCT04124926 / Laine et al.)Healing of Erosive Esophagitis (EE)3Randomized, Double-Blind, MulticenterVonoprazan 20 mg vs. Lansoprazole 30 mgEE healing rate at Week 8Vonoprazan was non-inferior and superior to lansoprazole (92.9% vs. 84.6%).7
PHALCON-EE (NCT04124926 / Laine et al.)Maintenance of Healed EE3Randomized, Double-Blind, MulticenterVonoprazan 10 mg vs. Lansoprazole 15 mgMaintenance of healing at 24 weeksVonoprazan was superior to lansoprazole (79.2% vs. 72.0%).7
PHALCON-HP (NCT04198363 / Chey et al.)H. pylori Eradication3Randomized, Partially Blinded, MulticenterVonoprazan Triple Therapy vs. Lansoprazole Triple TherapyEradication rate in clarithromycin-susceptible patientsVonoprazan Triple Therapy was non-inferior to Lansoprazole Triple Therapy (84.7% vs. 78.8%).7
PHALCON-HP (NCT04198363 / Chey et al.)H. pylori Eradication3Randomized, Partially Blinded, MulticenterVonoprazan Dual Therapy vs. Lansoprazole Triple TherapyEradication rate in clarithromycin-susceptible patientsVonoprazan Dual Therapy was non-inferior to Lansoprazole Triple Therapy (78.5% vs. 78.8%).7
Ashida et al. (2016)Healing of EE3Randomized, Double-Blind, Dose-RangingVonoprazan (5, 10, 20, 40 mg) vs. Lansoprazole 30 mgEE healing rate at Week 4All vonoprazan doses were non-inferior to lansoprazole. VPZ 20 mg and 40 mg were highly effective for severe EE.32
Kawai et al. (2018)Prevention of Low-Dose Aspirin-Induced Ulcers3Randomized, Double-BlindVonoprazan (10, 20 mg) vs. Lansoprazole 15 mgPeptic ulcer recurrence rate at 24 weeksVonoprazan was non-inferior to lansoprazole for ulcer prevention.46

VI. Safety, Tolerability, and Risk Management

The safety profile of vonoprazan has been characterized through an extensive program of clinical trials and post-marketing surveillance. While it is generally well-tolerated with a safety profile largely comparable to PPIs, its more potent mechanism raises considerations for both shared class effects and unique safety signals.

Common Adverse Reactions

Across clinical trials for various indications, vonoprazan has been shown to be well-tolerated. The most frequently reported adverse reactions (typically occurring in ≥2% of patients) are primarily gastrointestinal and generally mild to moderate in severity. These include gastritis, diarrhea, abdominal distension, abdominal pain, and nausea. Other commonly reported events include dyspepsia, hypertension, and urinary tract infection.[6]

Serious Adverse Reactions and Class-Effect Warnings

As a potent inhibitor of gastric acid secretion, vonoprazan shares several warnings and precautions with the PPI class, which are related to the long-term physiological consequences of profound acid suppression.

  • Clostridioides difficile-Associated Diarrhea (CDAD): The reduction of gastric acid, a natural barrier to enteric pathogens, can alter the gut microbiome and increase the risk of infections, most notably CDAD. This risk is a recognized class effect for all potent acid suppressants.[2]
  • Bone Fractures: Long-term use (typically over a year) and/or high doses of acid-suppressing agents have been associated with an increased risk of osteoporosis-related fractures of the hip, wrist, or spine. The mechanism is thought to involve reduced calcium absorption.[2]
  • Vitamin B12 (Cobalamin) Deficiency: Gastric acid is required to release vitamin B12 from food proteins. Prolonged and profound acid suppression can therefore lead to malabsorption and, eventually, deficiency of vitamin B12.[2]
  • Hypomagnesemia: Cases of low serum magnesium levels have been reported with prolonged treatment (over one year) with acid-suppressing drugs. This can be serious and may lead to tetany, arrhythmias, and seizures. The risk may be increased when taken with other drugs that can also lower magnesium, such as diuretics.[2]
  • Fundic Gland Polyps: Long-term use of acid suppressants is associated with the development of benign fundic gland polyps in the stomach, a consequence of elevated serum gastrin levels resulting from reduced acid feedback.[2]

Vonoprazan-Specific Safety Signals

In addition to the shared class effects, post-marketing experience, particularly from Japan where the drug has been used for the longest period, has identified some specific and serious safety signals for vonoprazan.

  • Severe Cutaneous Adverse Reactions (SCAR): Rare but life-threatening skin reactions, including Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), have been reported with vonoprazan use. Patients should be advised to discontinue the drug at the first sign of a severe rash.[44]
  • Acute Tubulointerstitial Nephritis (TIN): Cases of TIN, an acute kidney injury, have been reported. This is a hypersensitivity reaction that can occur at any time during treatment.[48]
  • Hemorrhagic Enterocolitis: A notable finding from a Japanese pharmacovigilance study was a strong statistical association between vonoprazan use and hemorrhagic enterocolitis, with a reporting odds ratio (ROR) of 86.5. This signal was not significantly detected for the PPI class, suggesting it may be a unique risk associated with vonoprazan.[55]

The emergence of these unique safety signals highlights a critical consideration in the clinical application of vonoprazan. While the drug was designed to solve the pharmacokinetic and pharmacodynamic limitations of PPIs, its very success in achieving more potent and sustained acid suppression may introduce new physiological consequences. The safety profile from short-term clinical trials appeared largely similar to that of the well-established PPIs, which was reassuring for initial regulatory approvals. However, the post-marketing data from Japan, with its longer real-world experience, revealed the potential for distinct and serious adverse events like hemorrhagic enterocolitis. This suggests that the more profound alteration of the gastric environment by P-CABs could lead to different downstream effects on the intestinal mucosa or microbiome compared to PPIs. Therefore, the long-term safety of vonoprazan cannot be entirely extrapolated from the decades of experience with PPIs. Clinicians and regulatory bodies must remain vigilant for P-CAB-specific adverse events as global use expands, continually weighing the established benefits of superior efficacy against an evolving and potentially distinct long-term risk profile.

Use in Specific Populations

  • Pregnancy and Lactation: There is a lack of sufficient human data on the use of vonoprazan during pregnancy. Animal reproductive toxicology studies have shown fetal abnormalities, but only at maternal exposures many times higher than the maximum recommended human dose.[27] To gather more data, a pregnancy exposure registry has been established to monitor outcomes in women exposed to vonoprazan during pregnancy. Due to the lack of safety data and findings of adverse effects in nursing pups in animal studies, breastfeeding is not recommended during treatment with vonoprazan.[27]
  • Renal and Hepatic Impairment: The clearance of vonoprazan can be affected by severe organ impairment. Dose adjustments are required for the healing of erosive esophagitis in patients with severe renal impairment (eGFR < 30 mL/min) or moderate to severe hepatic impairment (Child-Pugh Class B or C). For the treatment of H. pylori infection, the use of vonoprazan is not recommended in these populations, partly due to the complexities of adjusting the doses of the co-administered antibiotics.[34]

VII. Drug Interaction Profile: Metabolic and pH-Dependent Mechanisms

Vonoprazan possesses a significant potential for drug-drug interactions (DDIs), with analyses identifying over 240 interacting drugs of varying clinical significance.[52] These interactions stem from two principal mechanisms: the alteration of gastric pH, which affects the absorption of other drugs, and the modulation of CYP450 enzymes, which affects drug metabolism.

Mechanism 1: pH-Dependent Absorption Interactions

By potently and sustainably increasing intragastric pH, vonoprazan can significantly reduce the absorption and bioavailability of drugs that require an acidic environment for their dissolution and absorption. This can lead to reduced efficacy of the co-administered drug.

  • Clinically Significant Examples: This interaction is particularly critical for certain antiretroviral drugs, azole antifungals, and tyrosine kinase inhibitors.
  • Antiretrovirals: The absorption of rilpivirine is highly pH-dependent, and its co-administration with vonoprazan is contraindicated due to the risk of therapeutic failure and development of viral resistance. Similarly, the absorption of atazanavir and nelfinavir is significantly reduced, and co-administration should be avoided.[13]
  • Antifungals: The absorption of ketoconazole and itraconazole is impaired in an alkaline environment, potentially leading to treatment failure.[13]
  • Tyrosine Kinase Inhibitors: The bioavailability of several oral cancer drugs, such as erlotinib and dasatinib, is reduced, which can compromise their anti-neoplastic effects.[13]

Mechanism 2: Cytochrome P450 (CYP) Enzyme Interactions

Vonoprazan is both a substrate and an inhibitor of CYP enzymes, creating a bidirectional potential for metabolic DDIs.

  • Vonoprazan as a Substrate: As vonoprazan is primarily metabolized by CYP3A4, its plasma concentrations can be altered by strong modulators of this enzyme.[12]
  • CYP3A4 Inducers: Strong inducers of CYP3A4, such as rifampin, carbamazepine, and St. John's Wort, can significantly increase the metabolism of vonoprazan, leading to lower plasma levels and potentially reduced therapeutic efficacy. Concomitant use with strong inducers should be avoided.[35]
  • CYP3A4 Inhibitors: Strong inhibitors of CYP3A4, such as clarithromycin, can decrease the metabolism of vonoprazan, leading to increased plasma concentrations. This interaction is intentionally leveraged in the vonoprazan-based triple therapy for H. pylori eradication, as both drugs mutually inhibit each other's metabolism, boosting the exposure and efficacy of both agents.[12]
  • Vonoprazan as an Inhibitor: Vonoprazan itself can inhibit the metabolism of other drugs.
  • CYP3A4 Inhibition: It is considered a weak inhibitor of CYP3A4. While this effect is generally modest, caution is advised when it is co-administered with sensitive CYP3A4 substrates that have a narrow therapeutic index, such as alfentanil.[13]
  • CYP2C19 Inhibition: Vonoprazan is a clinically relevant inhibitor of CYP2C19. This has important implications for drugs that are substrates of this enzyme, such as clopidogrel and certain antidepressants. This interaction is discussed in greater detail in the subsequent section.

Notable Non-Interactions

Importantly, dedicated DDI studies have demonstrated that vonoprazan does not have any clinically meaningful pharmacokinetic interactions with low-dose aspirin or the commonly used NSAIDs loxoprofen, diclofenac, and meloxicam. This finding is crucial as it supports the safety of its use for the prevention of NSAID- and aspirin-induced peptic ulcers in patients who require these concomitant medications.[26]

The following table summarizes some of the most clinically significant drug interactions with vonoprazan, providing guidance for prescribers.

Table 4: Clinically Significant Drug-Drug Interactions with Vonoprazan

Interacting Drug / ClassMechanism of InteractionClinical Effect on Interacting DrugRecommendationSource(s)
RilpivirineIncreased gastric pH reduces absorptionDecreased plasma concentration and loss of virologic responseContraindicated34
Atazanavir, NelfinavirIncreased gastric pH reduces absorptionDecreased plasma concentration and loss of virologic responseAvoid concomitant use13
Ketoconazole, ItraconazoleIncreased gastric pH reduces absorptionDecreased plasma concentration and potential loss of efficacyAvoid concomitant use13
Iron SaltsIncreased gastric pH reduces absorptionDecreased iron absorptionAdminister separately if possible; monitor for efficacy13
Strong CYP3A4 Inducers (e.g., Rifampin, St. John's Wort)Increased metabolism of vonoprazanDecreased vonoprazan concentration and potential loss of efficacyAvoid concomitant use13
Strong CYP3A4 Inhibitors (e.g., Clarithromycin)Decreased metabolism of vonoprazanIncreased vonoprazan concentrationMonitor for adverse effects; interaction is utilized in H. pylori therapy12
CYP2C19 Substrates (e.g., Clopidogrel, Citalopram)Inhibition of CYP2C19 by vonoprazanIncreased concentration of active drug (citalopram) or decreased concentration of active metabolite (clopidogrel)Monitor for efficacy/toxicity; consider alternatives13
Sensitive CYP3A4 Substrates (e.g., Alfentanil, Midazolam)Weak inhibition of CYP3A4 by vonoprazanPotentially increased concentration of substrateUse with caution, monitor for adverse effects13

VIII. The CYP2C19 Question: A Nuanced Discussion

The relationship between vonoprazan and the cytochrome P450 enzyme CYP2C19 is a topic of critical pharmacological importance and clinical nuance. It represents both a key advantage over traditional PPIs and a source of potential drug-drug interactions, a duality that must be clearly understood for safe and effective prescribing.

Independence from Genotype: A Pharmacokinetic Advantage

A primary limitation of many widely used PPIs, such as omeprazole and lansoprazole, is their heavy reliance on CYP2C19 for metabolism. The gene for this enzyme is highly polymorphic, leading to significant inter-individual variability in drug clearance and clinical response.[5] In contrast, vonoprazan's metabolism is dominated by CYP3A4, with CYP2C19 playing only a minor role.[8]

The clinical consequence of this metabolic profile is that the pharmacokinetics and, therefore, the acid-suppressive effects of vonoprazan are not clinically significantly influenced by a patient's CYP2C19 genotype.[7] This means that the drug provides a consistent and predictable level of acid inhibition regardless of whether the patient is a poor, intermediate, or extensive metabolizer of CYP2C19. This reliability eliminates a major source of therapeutic failure seen with PPIs and removes any potential need for pharmacogenetic testing to guide dosing, simplifying its clinical application.[32]

Inhibition of CYP2C19: A Drug Interaction Liability

While vonoprazan's own disposition is largely independent of CYP2C19, a separate and crucial finding is that vonoprazan itself acts as a clinically relevant inhibitor of the CYP2C19 enzyme.[57] This has been demonstrated in dedicated drug interaction studies. For instance, a randomized crossover study in healthy volunteers used the probe drug proguanil, which is primarily metabolized by CYP2C19, to assess the enzyme's activity. The study found that co-administration of vonoprazan, much like the known CYP2C19 inhibitor esomeprazole, significantly decreased the metabolism of proguanil to its active metabolite, cycloguanil. This confirmed that vonoprazan exerts an inhibitory effect on CYP2C19

in vivo.[57]

This inhibitory action has direct clinical implications. It may be the mechanism underlying the reported attenuation of the antiplatelet effect of clopidogrel, a prodrug that requires activation by CYP2C19.[57] For patients taking clopidogrel, concomitant use of a CYP2C19 inhibitor like vonoprazan could lead to reduced platelet inhibition and an increased risk of thrombotic events. Similarly, vonoprazan could increase the plasma concentrations of other CYP2C19 substrates, such as the antidepressant citalopram or the anticonvulsant phenytoin, potentially increasing the risk of toxicity.

The relationship between vonoprazan and CYP2C19 thus presents a complex clinical picture that can be a source of confusion. The marketing and clinical positioning of the drug often highlight its "independence from CYP2C19" as a major advantage, which is true with respect to its own predictable efficacy. However, this message can inadvertently obscure the other side of the interaction: its potential to cause clinically significant DDIs by inhibiting the metabolism of other drugs. This distinction is subtle but critical. Vonoprazan solves the problem of variable patient response due to genetics, but it introduces a risk of altering the response to other medications through the very same enzyme. This underscores the necessity for clinicians to look beyond headline advantages and understand the full pharmacological profile of a drug. The safe use of vonoprazan, particularly in patients on polypharmacy, requires an appreciation of this duality: it offers reliable pharmacokinetics for itself but demands continued vigilance for its effects on co-administered CYP2C19 substrates.

IX. Regulatory Status, Dosing, and Clinical Positioning

Vonoprazan was discovered and developed by Takeda Pharmaceutical Company Limited, which first launched the drug in Japan, marking its entry as a novel therapeutic agent for acid-related disorders.[10] Recognizing its potential in Western markets, Phathom Pharmaceuticals subsequently licensed the exclusive rights for the development and commercialization of vonoprazan in the United States, Europe, and Canada.[39]

Brand Names and Global Approvals

Vonoprazan has achieved regulatory approval in numerous countries under various brand names, reflecting its growing global presence.

  • Japan: First approved by the Pharmaceuticals and Medical Devices Agency (PMDA) in December 2014 and launched in February 2015 under the brand name Takecab®.[12] It is co-promoted in Japan by Takeda and Otsuka Pharmaceutical.[59]
  • United States: Marketed by Phathom Pharmaceuticals under the brand name Voquezna®. The U.S. Food and Drug Administration (FDA) first approved co-packaged products for H. pylori eradication (Voquezna Triple Pak and Voquezna Dual Pak) in May 2022.[18] This was followed by the approval of vonoprazan monotherapy for the healing and maintenance of Erosive GERD in November 2023, and an expanded indication for the treatment of heartburn associated with Non-Erosive GERD in July 2024.[18]
  • Europe: As of August 2024, vonoprazan has not yet received a full marketing authorization from the European Medicines Agency (EMA). However, the regulatory process is underway, as evidenced by an agreed Paediatric Investigation Plan (PIP), which is a mandatory step before a marketing authorization application can be finalized and submitted for review.[62] The brand name for the European market has not been officially confirmed.
  • Other Regions: Vonoprazan has also been approved in other markets, including Russia (as Vocinti) and several other countries in Asia and Latin America.[18]

Clinical Positioning and Market Impact

Vonoprazan is strategically positioned as a premium, next-generation acid suppressant that offers distinct advantages over the long-standing PPI class. Its approval in the U.S. was hailed as the first major innovation in the Erosive GERD market in over three decades, addressing the significant unmet needs of patients dissatisfied with current therapies.[31]

Its clinical positioning is built on several key pillars:

  1. Superior Efficacy: Particularly in more severe grades of erosive esophagitis and in the eradication of H. pylori, where it has demonstrated higher success rates than PPIs, especially against antibiotic-resistant strains.[4]
  2. Reliability and Predictability: By circumventing the issue of CYP2C19 genetic polymorphism, vonoprazan offers a more consistent and reliable therapeutic effect across diverse patient populations.[7]
  3. Patient Convenience: The ability to be dosed without regard to meals offers a significant lifestyle advantage and can improve treatment adherence compared to the strict pre-meal regimen of PPIs.[6]

Approved Dosing Regimens

The recommended adult dosages for vonoprazan vary by indication and regulatory jurisdiction. The following table provides a comparative summary of the approved regimens in the United States (FDA) and Japan (PMDA).

Table 5: Approved Adult Dosing Regimens for Vonoprazan by Indication and Regulatory Agency

IndicationRegulatory AgencyRecommended Adult DosageSource(s)
Healing of Erosive EsophagitisFDA (USA)20 mg once daily for 8 weeks2
PMDA (Japan)20 mg once daily for up to 4 weeks (may be extended to 8 weeks if response is inadequate)29
Maintenance of Healed Erosive EsophagitisFDA (USA)10 mg once daily for up to 6 months2
PMDA (Japan)10 mg once daily (may be increased to 20 mg once daily if response is inadequate)59
Heartburn associated with Non-Erosive GERDFDA (USA)10 mg once daily for 4 weeks34
PMDA (Japan)Not explicitly listed as a standalone indication in provided documents.43
H. pylori Eradication (Triple Therapy)FDA (USA)Vonoprazan 20 mg BID + Amoxicillin 1000 mg BID + Clarithromycin 500 mg BID for 14 days35
PMDA (Japan)Vonoprazan 20 mg BID + Amoxicillin 750 mg BID + Clarithromycin 200 mg BID (may be increased to 400 mg BID) for 7 days29
H. pylori Eradication (Dual/Alternative Therapy)FDA (USA)Vonoprazan 20 mg BID + Amoxicillin 1000 mg TID for 14 days35
PMDA (Japan)For second-line therapy: Vonoprazan 20 mg BID + Amoxicillin 750 mg BID + Metronidazole 250 mg BID for 7 days29
Prevention of NSAID-Induced Ulcer RecurrenceFDA (USA)Not an approved indication.49
PMDA (Japan)10 mg once daily29
Prevention of Aspirin-Induced Ulcer RecurrenceFDA (USA)Not an approved indication.49
PMDA (Japan)10 mg once daily29

Note: BID = twice daily; TID = three times daily.

X. Conclusion

Vonoprazan represents a landmark achievement in the field of gastroenterology, establishing the potassium-competitive acid blocker (P-CAB) class as a formidable successor to proton pump inhibitors (PPIs). Its development was a direct response to the well-defined pharmacological and clinical limitations of PPIs, and its profile demonstrates a successful translation of rational drug design into tangible patient benefits.

Pharmacologically, vonoprazan's mechanism of direct, reversible, and competitive inhibition of the H+/K+-ATPase, combined with its acid stability and high pKa, results in a more rapid, potent, and durable acid suppression than is achievable with PPIs. This superior pharmacodynamic profile is supported by a convenient pharmacokinetic profile, characterized by a long half-life, predictable behavior, and an absence of clinically significant food effects or variability due to CYP2C19 genetic polymorphisms.

In the clinical arena, these pharmacological advantages have translated into compelling efficacy. Vonoprazan has demonstrated superiority over standard-of-care PPIs in the healing of severe erosive esophagitis and in the long-term maintenance of remission. Furthermore, it has redefined the approach to H. pylori eradication, offering higher cure rates, particularly against clarithromycin-resistant strains, and providing novel, effective dual-therapy options.

However, the clinical profile of vonoprazan is not without complexities. While its short-term safety is comparable to that of PPIs, its more profound acid suppression may introduce a distinct long-term risk profile, as suggested by post-marketing signals of rare but serious adverse events like hemorrhagic enterocolitis. Additionally, its role as a clinically relevant inhibitor of the CYP2C19 enzyme necessitates careful consideration of drug-drug interactions, a nuance that must be clearly communicated to prescribers.

In conclusion, vonoprazan is a transformative therapeutic agent that offers significant advantages in efficacy, reliability, and convenience for the management of acid-related disorders. It has rightfully earned its place as a first-line option for severe GERD and as a superior component of H. pylori eradication regimens. As its use expands globally, ongoing pharmacovigilance and further research into its long-term safety will be essential to fully define its role and ensure its benefits continue to be weighed appropriately against its potential risks, solidifying its position as a cornerstone of modern acid suppression therapy.

Works cited

  1. What is the mechanism of Vonoprazan Fumarate? - Patsnap Synapse, accessed August 3, 2025, https://synapse.patsnap.com/article/what-is-the-mechanism-of-vonoprazan-fumarate
  2. Vonoprazan: Digestive Uses, Side Effects, Dosage - MedicineNet, accessed August 3, 2025, https://www.medicinenet.com/vonoprazan/article.htm
  3. Progress in the Study of Vonoprazan Fumarate vs. Proton Pump Inhibitors in the Treatment of Gastroesophageal Reflux Disease - Scientific Research Publishing, accessed August 3, 2025, https://www.scirp.org/journal/paperinformation?paperid=125511
  4. Vonoprazan and proton pump inhibitors: Which is superior for ..., accessed August 3, 2025, https://www.wjgnet.com/1007-9327/full/v31/i17/103156.htm
  5. Association between CYP2C19 polymorphism and proton pump inhibitors adverse drug reactions: a narrative review - Frontiers, accessed August 3, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2025.1523399/full
  6. What is the mechanism of action of Vonoprazan (Proton Pump Inhibitor) PPI? - Dr.Oracle, accessed August 3, 2025, https://www.droracle.ai/articles/2262/what-is-the-mechanism-of-action-of-voquezna-
  7. Vonoprazan: A New Potassium-Competitive Acid Blocker - PMC, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10268044/
  8. (PDF) The Effect of Food on the Pharmacokinetics of the Potassium‐Competitive Acid Blocker Vonoprazan - ResearchGate, accessed August 3, 2025, https://www.researchgate.net/publication/354123241_The_Effect_of_Food_on_the_Pharmacokinetics_of_the_Potassium-Competitive_Acid_Blocker_Vonoprazan
  9. Vonoprazan in Management of Refractory Gastroesophageal Reflux Disease: An Indian Expert Group Consensus Statements - JAPI, accessed August 3, 2025, https://www.japi.org/article/japi-73-3-e29
  10. CAS 881681-00-1 Vonoprazan - BOC Sciences, accessed August 3, 2025, https://www.bocsci.com/vonoprazan-cas-881681-00-1-item-474886.html
  11. Vonoprazan for treatment of gastroesophageal reflux: pharmacodynamic and pharmacokinetic considerations - CORE, accessed August 3, 2025, https://core.ac.uk/download/pdf/54924292.pdf
  12. The First-in-Class Potassium-Competitive Acid Blocker, Vonoprazan Fumarate: Pharmacokinetic and Pharmacodynamic Considerations - PubMed, accessed August 3, 2025, https://pubmed.ncbi.nlm.nih.gov/26369775/
  13. Vonoprazan: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 3, 2025, https://go.drugbank.com/drugs/DB11739
  14. Effect of CYP2C19 Genetic Polymorphism on... : The Journal of Clinical Pharmacology, accessed August 3, 2025, https://www.ovid.com/journals/jclph/abstract/10.1177/0091270011408611~effect-of-cyp2c19-genetic-polymorphism-on-pharmacokinetics
  15. Vonoprazan fumarate - TOKU-E, accessed August 3, 2025, https://toku-e.com/vonoprazan-fumarate/
  16. CAS No : 881681-00-1| Chemical Name : Vonoprazan - Pharmaffiliates, accessed August 3, 2025, https://www.pharmaffiliates.com/en/881681-00-1-vonoprazan-api-pa2247000.html
  17. Vonoprazan fumarate, a novel potassium-competitive acid blocker, in the management of gastroesophageal reflux disease: safety and clinical evidence to date - PMC - PubMed Central, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5784563/
  18. Vonoprazan - Wikipedia, accessed August 3, 2025, https://en.wikipedia.org/wiki/Vonoprazan
  19. S-1/A - SEC.gov, accessed August 3, 2025, https://www.sec.gov/Archives/edgar/data/1783183/000119312519267053/d63338ds1a.htm
  20. Compound: VONOPRAZAN FUMARATE (CHEMBL2064032) - ChEMBL - EMBL-EBI, accessed August 3, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL2064032
  21. Vonoprazan | C17H16FN3O2S - ChemSpider, accessed August 3, 2025, https://www.chemspider.com/Chemical-Structure.13112797.html
  22. Vonoprazan | C17H16FN3O2S | CID 15981397 - PubChem, accessed August 3, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Vonoprazan
  23. Vonoprazan | 881681-00-1 - Sigma-Aldrich, accessed August 3, 2025, https://www.sigmaaldrich.com/US/en/product/ambeedinc/ambh2d6ff9ac?context=bbe
  24. Pharmacokinetic Comparison and Bioequivalence Evaluation of Two 20-mg Vonoprazan Fumarate Tablets in Bangladeshi Healthy Male Subjects, accessed August 3, 2025, https://www.scirp.org/journal/paperinformation?paperid=135401
  25. A Population Pharmacokinetic Model of Vonoprazan: Evaluating the Effects of Race, Disease Status, and Other Covariates on Exposure - PubMed Central, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9303766/
  26. Pharmacokinetic Drug–Drug Interactions Between Vonoprazan and Low-Dose Aspirin or Nonsteroidal Anti-inflammatory Drugs: A Phase 2, Open-Label, Study in Healthy Japanese Men, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5209422/
  27. APPLICATION NUMBER: - 215151Orig1s000 OTHER REVIEW(S) - accessdata.fda.gov, accessed August 3, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2024/215151Orig1s000OtherR.pdf
  28. (PDF) A Population Pharmacokinetic Model of Vonoprazan: Evaluating the Effects of Race, Disease Status, and Other Covariates on Exposure - ResearchGate, accessed August 3, 2025, https://www.researchgate.net/publication/357254661_A_Population_Pharmacokinetic_Model_of_Vonoprazan_Evaluating_the_Effects_of_Race_Disease_Status_and_Other_Covariates_on_Exposure
  29. VOCINTI - National Drug Information, accessed August 3, 2025, https://ndi.fda.moph.go.th/uploads/drug_detail_corporation/doc/word/1163/59617143799c78416f40a1fd7d1442e5-a1.pdf
  30. Study Details | Efficacy and Safety of Vonoprazan Compared to Lansoprazole in Participants With Erosive Esophagitis | ClinicalTrials.gov, accessed August 3, 2025, https://clinicaltrials.gov/study/NCT04124926
  31. FDA Approves Vonoprazan for the Treatment of Erosive Esophagitis, accessed August 3, 2025, https://www.pharmexec.com/view/fda-approves-vonoprazan-for-the-treatment-of-erosive-esophagitis
  32. Randomised clinical trial: a dose‐ranging study of vonoprazan, a ..., accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5014135/
  33. Phathom Pharmaceuticals Announces FDA Approval of VOQUEZNA® (vonoprazan) Tablets for the Treatment of Erosive GERD and Relief of Heartburn Associated with Erosive GERD in Adults, accessed August 3, 2025, https://investors.phathompharma.com/news-releases/news-release-details/phathom-pharmaceuticals-announces-fda-approval-voqueznar/
  34. Vonoprazan (oral route) - Side effects & dosage - Mayo Clinic, accessed August 3, 2025, https://www.mayoclinic.org/drugs-supplements/vonoprazan-oral-route/description/drg-20559712
  35. Voquezna (vonoprazan) dosing, indications, interactions, adverse effects, and more, accessed August 3, 2025, https://reference.medscape.com/drug/voquezna-vonoprazan-4000266
  36. What is Vonoprazan? - Dr.Oracle AI, accessed August 3, 2025, https://www.droracle.ai/articles/4518/tell-me-about-voquezna-
  37. VOQUEZNA TRIPLE PAK (vonoprazan - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed August 3, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/215152s000,215153s000lbl.pdf
  38. 215152Orig1s000 215153Orig1s000 - accessdata.fda.gov, accessed August 3, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2022/215152Orig1s000,215153Orig1s000OtherR.pdf
  39. VOQUEZNA (vonoprazan) for the Treatment of Helicobacter Pylori Infection, USA, accessed August 3, 2025, https://www.clinicaltrialsarena.com/projects/voquezna-vonoprazan-h-pylori-infection/
  40. Vonoprazan Completed Phase 3 Trials for Helicobacter Pylori Treatment | DrugBank Online, accessed August 3, 2025, https://go.drugbank.com/drugs/DB11739/clinical_trials?conditions=DBCOND0034525&phase=3&purpose=treatment&status=completed
  41. The safety and effectiveness of vonoprazan-based Helicobacter pylori eradication therapy; a prospective post-marketing surveillance - Taylor & Francis Online, accessed August 3, 2025, https://www.tandfonline.com/doi/full/10.1080/14740338.2019.1676722
  42. Vonoprazan versus proton pump inhibitors for the management of, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6181618/
  43. Summary of Investigation Results - PMDA, accessed August 3, 2025, https://www.pmda.go.jp/files/000236942.pdf
  44. Preparations containing vonoprazan fumarate - PMDA, accessed August 3, 2025, https://www.pmda.go.jp/files/000228668.pdf
  45. New Drug Application Approval of TAKECAB® for the treatment of Acid-related Diseases in Japan - Takeda Pharmaceuticals, accessed August 3, 2025, https://www.takeda.com/newsroom/newsreleases/2014/new-drug-application-approval-of-takecab-for-the-treatment-of-acid-related-diseases-in-japan/
  46. Vonoprazan in the management of erosive oesophagitis and peptic ulcer-induced medication: a systematic review - PMC, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9475477/
  47. Vonoprazan in the management of gastric/peptic ulcers: a systematic review of safety data, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9743326/
  48. VOQUEZNA® (vonoprazan) for GERD and H. pylori Infection in adults | HCP, accessed August 3, 2025, https://voqueznapro.com/
  49. Vonoprazan - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed August 3, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/215151s000lbl.pdf
  50. Phathom Pharmaceuticals Launches Direct-to-Consumer Campaign, “VOQUEZNA Can Kick Some Acid”, accessed August 3, 2025, https://investors.phathompharma.com/news-releases/news-release-details/phathom-pharmaceuticals-launches-direct-consumer-campaign/
  51. Vonoprazan: MedlinePlus Drug Information, accessed August 3, 2025, https://medlineplus.gov/druginfo/meds/a624006.html
  52. Vonoprazan Interactions Checker - Drugs.com, accessed August 3, 2025, https://www.drugs.com/drug-interactions/vonoprazan.html
  53. Voquezna Interactions Checker - Drugs.com, accessed August 3, 2025, https://www.drugs.com/drug-interactions/vonoprazan,voquezna.html
  54. 215151Orig1s000 INTEGRATED REVIEW - accessdata.fda.gov, accessed August 3, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2024/215151Orig1s000IntegratedR.pdf
  55. Safety profile of vonoprazan compared with proton pump inhibitors: insight from a pharmacovigilance study | Request PDF - ResearchGate, accessed August 3, 2025, https://www.researchgate.net/publication/348408683_Safety_profile_of_vonoprazan_compared_with_proton_pump_inhibitors_insight_from_a_pharmacovigilance_study
  56. VOQUEZNA® Tablets Prescriber Information | Phathom Pharmaceuticals, accessed August 3, 2025, https://www.phathompharma.com/wp-content/uploads/VOQUEZNA-tablets-Prescriber-Information.pdf
  57. (PDF) Evaluation of CYP2C19-Mediated Pharmacokinetic Drug Interaction of Tegoprazan, Compared with Vonoprazan or Esomeprazole - ResearchGate, accessed August 3, 2025, https://www.researchgate.net/publication/369145582_Evaluation_of_CYP2C19-Mediated_Pharmacokinetic_Drug_Interaction_of_Tegoprazan_Compared_with_Vonoprazan_or_Esomeprazole
  58. Evaluation of CYP2C19-Mediated Pharmacokinetic Drug Interaction of Tegoprazan, Compared with Vonoprazan or Esomeprazole - PMC, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10085907/
  59. TAKECAB® Now Available for the Treatment of Acid-related Diseases in Japan, accessed August 3, 2025, https://www.takeda.com/newsroom/newsreleases/2015/takecab-now-available-for-the-treatment-of-acid-related-diseases-in-japan/
  60. Evonik and Phathom Pharmaceuticals partner to produce novel acid-blocker vonoprazan, accessed August 3, 2025, https://healthcare.evonik.com/en/evonik-and-phathom-pharmaceuticals-partner-to-produce-novel-acid-blocker-vonoprazan-178267.html
  61. Evonik and Phathom Pharmaceuticals partner on novel acid-blocker vonoprazan, accessed August 3, 2025, https://www.labiotech.eu/trends-news/evonik-phathom-pharmaceuticals-partner-acid-blocker-vonoprazan/
  62. EMEA-002703-PIP01-19-M01 - paediatric investigation plan - EMA - European Union, accessed August 3, 2025, https://www.ema.europa.eu/en/medicines/human/paediatric-investigation-plans/emea-002703-pip01-19-m01
  63. European Medicines Agency decision P/0277/2024 of 14 August 2024 on the acceptance of a modification of an agreed paediatric inv - EMA, accessed August 3, 2025, https://www.ema.europa.eu/en/documents/pip-decision/p-0277-2024-ema-decision-14-august-2024-acceptance-modification-agreed-paediatric-investigation-plan-vonoprazan-emea-002703-pip01-19-m01_en.pdf
  64. pmc.ncbi.nlm.nih.gov, accessed August 3, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6181618/#:~:text=The%20acid%2Dinhibitory%20effect%20of,low%2Ddose%2Daspirin%20ulcer.
  65. Report on the Deliberation Results November 26, 2014 Evaluation and Licensing Division, Pharmaceutical and Food Safety Bureau Mi - PMDA, accessed August 3, 2025, https://www.pmda.go.jp/files/000211075.pdf
  66. Takecab Tablets 10mg [agents for digestive organ] | Kusuri-no-Shiori(Drug Information Sheet), accessed August 3, 2025, https://www.rad-ar.or.jp/siori/english/search/result?n=35520
  67. Study Details | Drug Use Surveillance of Takecab for "Supplement to Helicobacter Pylori Eradication" | ClinicalTrials.gov, accessed August 3, 2025, https://clinicaltrials.gov/study/NCT03219723

Published at: August 3, 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.