MedPath

Vernakalant Advanced Drug Monograph

Published:Sep 13, 2025

Generic Name

Vernakalant

Brand Names

Brinavess

Drug Type

Small Molecule

Chemical Formula

C20H31NO4

CAS Number

794466-70-9

Associated Conditions

Atrial Fibrillation

Vernakalant (DB06217): A Comprehensive Pharmacological and Clinical Monograph

Drug Profile and Executive Summary

Vernakalant is a novel, small-molecule antiarrhythmic agent developed for the rapid pharmacological cardioversion of recent-onset atrial fibrillation (AF).[1] Classified as an atypical Class III antiarrhythmic, its mechanism of action is distinguished by a relatively atrial-selective blockade of multiple cardiac ion channels, which underlies both its therapeutic efficacy and its specific safety profile.[1] The primary indication for its intravenous formulation, marketed as Brinavess, is the conversion of recent-onset AF to normal sinus rhythm in specific adult populations: non-surgery patients with AF of seven days or less duration, and post-cardiac surgery patients with AF of three days or less duration.[1]

The pharmacodynamic profile of vernakalant is characterized by a multi-channel blockade that preferentially targets atrial tissue. It potently inhibits atrial-specific potassium currents, such as the ultra-rapid delayed rectifier current (IKur​) and the acetylcholine-activated potassium current (IKACh​), which prolongs the atrial action potential and refractory period.[1] Crucially, this is combined with a frequency-dependent blockade of sodium channels, an effect that is enhanced at the rapid heart rates characteristic of AF, thereby slowing conduction in a pathology-selective manner.[2] This unique combination of actions allows for rapid termination of the reentrant circuits that sustain AF, with minimal effects on ventricular repolarization, theoretically reducing the risk of Torsade de Pointes associated with other Class III agents.[1]

Clinical trials have consistently demonstrated the rapid efficacy of intravenous vernakalant. Pivotal studies, including ACT I and ACT III, showed conversion rates of approximately 51% within 90 minutes, compared to approximately 4% for placebo.[7] The head-to-head AVRO trial further established its superiority over the standard-of-care agent amiodarone in achieving rapid cardioversion, with a median time to conversion of approximately 11 minutes in responders.[9]

Despite this proven efficacy, the clinical use of vernakalant is strictly defined by significant safety considerations. The most prominent adverse events are hemodynamic, including hypotension and bradycardia, which are more frequent in patients with underlying heart failure.[2] Although rare, serious events including ventricular arrhythmia and a case of fatal cardiogenic shock have been reported and have heavily influenced its regulatory assessment.[12]

This complex risk-benefit profile has led to a notable divergence in global regulatory status. While vernakalant is approved for use in the European Union, Canada, and over 40 other countries, the United States Food and Drug Administration (FDA) has repeatedly declined to approve the drug, citing unresolved safety concerns.[12] Consequently, vernakalant remains a potent but niche therapeutic option, whose application is reserved for carefully selected patients in highly monitored clinical settings.

Molecular Identity and Physicochemical Properties

Vernakalant is classified as a small molecule drug, specifically an alcohol and a member of the phenol chemical class.[1] For clinical and research applications, it is often formulated as Vernakalant Hydrochloride, a salt form that confers enhanced aqueous solubility and stability compared to the free base.[16] The development of this agent has been complex, involving multiple corporate entities over time, including Cardiome Pharma, Astellas Pharma, and Merck, which is reflected in its various code designations such as RSD1235 and MK-6621.[14] This protracted development history is indicative of the significant clinical and regulatory challenges encountered, particularly related to the drug's safety profile, which has shaped its path to market.

The chemical structure of vernakalant consists of several key functional moieties: a 3,4-dimethoxyphenyl group connected via an ethoxy linker to a (1R,2R)-cyclohexyl ring, which in turn is substituted with a (3R)-pyrrolidin-3-ol group.[1] The precise stereochemistry of the molecule is critical for its biological activity. The International Union of Pure and Applied Chemistry (IUPAC) name for the free base is (3R)-1-cyclohexyl]pyrrolidin-3-ol.[1]

The molecular formula of the vernakalant free base is C20​H31​NO4​, corresponding to an average molecular weight of approximately 349.47 g/mol and a monoisotopic weight of 349.2253 Da.[1] The hydrochloride salt has the molecular formula

C20​H32​ClNO4​, with an average molecular weight of approximately 385.9 g/mol.[17] A comprehensive list of its various chemical and regulatory identifiers is provided in Table 2.1.

Table 2.1: Chemical and Regulatory Identifiers for Vernakalant

IdentifierValue (Vernakalant Free Base)Value (Vernakalant Hydrochloride)Source(s)
DrugBank IDDB06217DBSALT002376 (Salt)1
CAS Number794466-70-9748810-28-81
IUPAC Name(3R)-1-cyclohexyl]pyrrolidin-3-ol(3R)-1-cyclohexyl]pyrrolidin-3-ol hydrochloride1
Molecular FormulaC20​H31​NO4​C20​H32​ClNO4​1
Average Weight349.471 g/mol385.93 g/mol1
Monoisotopic Weight349.225308482 Da385.2019862 Da1
UNII9G468C8B137G4J1ZD9UQ3
ChEMBL IDCHEMBL2111112CHEMBL21073833
PubChem CID993004999300483
SynonymsRSD1235, Kynapid, Brinavess, Vemakalant, vernakalantumRSD1235 hydrochloride, Brinavess, Vernakalant HCl1
ATC CodeC01BG11-3

Pharmacodynamics and Mechanism of Action

Vernakalant exerts its antiarrhythmic effect through a complex and relatively unique mechanism involving the blockade of multiple cardiac ion channels, with a preferential effect on atrial tissue.[1] This action targets key electrophysiological processes in all phases of the atrial action potential, leading to a prolongation of the atrial effective refractory period (ERP) and a rate-dependent slowing of impulse conduction, which are critical for terminating the reentrant electrical activity that sustains atrial fibrillation.[1] While often categorized as a Class III antiarrhythmic, its potent effects on sodium channels give it significant Class I characteristics, making it a hybrid multi-channel blocker that does not fit neatly into the traditional Vaughan Williams classification.[1] This hybrid nature helps explain both its rapid onset of action, a feature more typical of Class I drugs, and its effects on repolarization.

Multi-Channel Blockade

The therapeutic action of vernakalant arises from its ability to modulate several distinct ion currents.

Potassium (K+) Channel Blockade

The blockade of specific potassium channels is the primary mechanism by which vernakalant prolongs atrial repolarization and refractoriness, a hallmark of Class III antiarrhythmic activity. Its atrial selectivity is largely derived from its affinity for K+ channels that are predominantly expressed in the atria.

  • Kv1.5 (IKur​): Vernakalant is a potent blocker of the voltage-gated potassium channel Kv1.5, which conducts the ultra-rapid delayed rectifier current (IKur​).[1] With a half-maximal inhibitory concentration ( IC50​) of 13.0 µM, this is one of its key interactions.[23] Since IKur​ is a major repolarizing current in human atria but is functionally absent in the ventricles, its blockade is a principal contributor to the drug's atrial-selective action.[1]
  • Kv4.3 (Ito​): The drug also blocks the Kv4.3 channel, responsible for the transient outward potassium current (Ito​), with an IC50​ of 30.0 µM.[1] Ito​ contributes to the early phase of repolarization and is more prominent in atrial than in ventricular tissue, further enhancing the drug's atrial-predominant effects.[1]
  • Kir3.1/Kir3.4 (IKACh​): Vernakalant inhibits the G-protein-coupled inwardly-rectifying potassium channels Kir3.1/Kir3.4, which conduct the acetylcholine-activated potassium current (IKACh​).[1] This current is activated by parasympathetic (vagal) stimulation, which shortens the atrial action potential and is a known trigger for AF. Inhibition of IKACh​ counteracts this effect and contributes to the prolongation of atrial refractoriness.[1]
  • KCNH2 (hERG/IKr​): A defining feature of vernakalant's intended safety profile is its minimal effect on the hERG channel, which conducts the rapid component of the delayed rectifier potassium current (IKr​).[1] The IC50​ for hERG is 21.0 µM, substantially higher than for Kv1.5.[23] Potent blockade of IKr​ is the primary cause of drug-induced QT prolongation and the life-threatening arrhythmia Torsade de Pointes. By largely sparing this channel, vernakalant produces only mild QT prolongation that resolves quickly after infusion, differentiating it from many other Class III agents and contributing to its low intrinsic risk of this specific proarrhythmia.[2]

Sodium (Na+) Channel Blockade

In addition to its effects on potassium channels, vernakalant exhibits a potent and clinically crucial blockade of voltage-gated sodium channels (Nav1.5, encoded by SCN5A), which is a characteristic of Class I antiarrhythmics.[1]

  • Potency: With an IC50​ of 9.0 µM, the blockade of Nav1.5 is the most potent of vernakalant's known ion channel interactions.[23] This action inhibits the rapid influx of sodium during phase 0 of the action potential, thereby slowing conduction velocity.
  • Frequency-Dependence: The most distinctive feature of its sodium channel blockade is its frequency-dependence (also known as use-dependence).[1] The affinity of vernakalant for the sodium channel increases significantly as the heart rate increases.[2] During the rapid and chaotic activation of AF, the channels are frequently in the open or inactivated states, to which vernakalant binds preferentially. This "pathology-selective" mechanism enhances the drug's effect precisely when it is needed most—in fibrillating atrial tissue. As the heart rate slows upon conversion to sinus rhythm, the drug rapidly unbinds, and its effect diminishes.[1]

The Principle of Atrial Selectivity

The preferential action of vernakalant on the atria is not due to a single mechanism but is rather the synergistic result of several properties. First, it targets ion channels (IKur​, IKACh​) that are functionally specific to or predominantly expressed in atrial myocytes.[1] Second, it exploits the unique electrophysiological environment of fibrillating atria—namely, a more depolarized resting membrane potential and extremely rapid activation rates—which enhances its state-dependent blockade of sodium channels.[1] Third, its relative sparing of the ventricular

IKr​ current minimizes its impact on ventricular repolarization.[1]

However, this atrial selectivity is relative, not absolute. The potent sodium channel blockade, which is central to its efficacy, is also the likely source of its most significant dose-limiting toxicity. While the effect is more pronounced in the atria, the blockade of sodium channels in the ventricles can still occur, leading to negative inotropy (reduced contractility) and slowed ventricular conduction. In vulnerable patients, particularly those with pre-existing systolic dysfunction or severe outflow obstruction, this effect can manifest as clinically significant hypotension and, in rare cases, cardiogenic shock.[12] This direct link between the drug's most potent therapeutic mechanism and its most dangerous adverse effect is fundamental to understanding its narrow therapeutic window and the stringent contraindications for its use.

Pharmacokinetics, Metabolism, and Excretion

The pharmacokinetic profile of vernakalant is characterized by its intravenous route of administration, rapid distribution, metabolism dependent on genetic polymorphism, and a relatively short elimination half-life, which are all well-suited for its use as an acute-care therapeutic agent.[2]

Administration and Distribution

Vernakalant is administered exclusively as an intravenous infusion over a 10-minute period; it is not intended for administration as an intravenous push or bolus.[11] This method ensures complete bioavailability and allows for precise dose control, with the ability to immediately terminate the infusion if adverse hemodynamic effects occur. Following administration, the drug undergoes rapid and extensive distribution throughout the body.[2] It exhibits low to moderate plasma protein binding, with the unbound (free) fraction in human serum ranging from 53% to 63%.[1] This high free fraction means a substantial portion of the administered dose is pharmacologically active and available to interact with its target ion channels.

Metabolism and Genetic Polymorphism

The primary route of elimination for vernakalant is hepatic metabolism, a process that is critically influenced by a common genetic polymorphism in the cytochrome P450 enzyme system.[1]

  • CYP2D6 Extensive Metabolizers (EMs): For the majority of the population who possess normal CYP2D6 enzyme activity, vernakalant is predominantly cleared via O-demethylation mediated by this enzyme.[1]
  • CYP2D6 Poor Metabolizers (PMs): In the subset of the population with deficient or absent CYP2D6 activity (approximately 7-10% of Caucasians), the metabolic pathway shifts. In these individuals, glucuronidation becomes the primary mechanism of metabolism.[1]

This difference in metabolic pathways based on genotype has a direct and significant impact on the drug's elimination rate and duration of action.

Elimination and Half-Life

Following metabolism, the metabolites of vernakalant are primarily eliminated from the body via renal excretion.[1] The drug's elimination half-life (

T1/2​) is directly dependent on the patient's CYP2D6 metabolizer status. In extensive metabolizers, the elimination half-life is approximately 3 hours.[1] In contrast, poor metabolizers exhibit a significantly longer elimination half-life of approximately 5.5 hours.[1] The typical total body clearance of vernakalant has been estimated to be 0.41 L/hr/kg.[1]

This nearly twofold difference in half-life between EMs and PMs has important clinical implications. While no specific dose adjustments are recommended based on genotype, the prolonged exposure in poor metabolizers suggests a longer duration of potential risk for adverse events such as hypotension and bradycardia. If such an event were to occur in a PM patient, its resolution might be delayed compared to an EM patient, adding a layer of pharmacokinetic variability to the drug's overall risk profile.

Table 4.1: Key Pharmacokinetic Parameters of Vernakalant

ParameterValuePopulation / ConditionSource(s)
Administration RouteIntravenous InfusionAll14
Protein Binding47-37% (53-63% free fraction)Human Serum1
Primary MetabolismCYP2D6 O-demethylation (EMs); Glucuronidation (PMs)Hepatic1
Elimination Half-Life (T1/2​)~3 hoursCYP2D6 Extensive Metabolizers (EMs)1
Elimination Half-Life (T1/2​)~5.5 hoursCYP2D6 Poor Metabolizers (PMs)1
Total Body Clearance0.41 L/hr/kgTypical1
Cmax​ (Single IV Dose)2.98 µg/mL3 mg/kg dose in Homo sapiens23
AUC (Single IV Dose)0.686 µg·h/mL0.5 mg/kg dose in Homo sapiens23

Clinical Efficacy and Trial Data Analysis

The clinical development program for intravenous vernakalant has robustly established its efficacy for the rapid conversion of recent-onset AF. The program was built upon several pivotal Phase III trials, including placebo-controlled studies and a key active-comparator trial, which collectively defined its therapeutic profile. Additionally, investigations into an oral formulation for the prevention of AF recurrence were conducted, though this formulation did not proceed to market.[28]

Pivotal Placebo-Controlled Trials (ACT I & ACT III)

The Atrial Arrhythmia Conversion Trials (ACT) I and III were foundational in demonstrating the efficacy of vernakalant. These were large, randomized, double-blind, placebo-controlled studies involving patients with symptomatic, recent-onset AF (duration of 3 hours to 7 days).[7] The primary efficacy endpoint in these trials was the proportion of patients who converted from AF to sinus rhythm for at least one minute within 90 minutes of the start of the drug infusion.[13]

The results were statistically significant and clinically compelling. In the ACT I trial, 51.7% of patients treated with vernakalant achieved the primary endpoint, compared to only 4.0% of patients in the placebo group (p<0.0001).[7] The ACT III trial produced nearly identical results, with a conversion rate of 51.2% in the vernakalant arm versus 3.6% in the placebo arm (

p<0.0001).[7] A key secondary finding across these studies was the remarkable speed of action; for patients who responded, the median time to conversion was between 8 and 11 minutes.[8]

Active-Comparator Trial (AVRO)

To establish the clinical utility of vernakalant relative to existing therapies, the AVRO (A Vernakalant vs. Amiodarone) trial was conducted.[9] This randomized, double-blind, head-to-head superiority trial compared intravenous vernakalant to intravenous amiodarone, a widely used but slower-acting antiarrhythmic, in patients with recent-onset AF (duration 3 to 48 hours).[10] The primary endpoint was again conversion to sinus rhythm within 90 minutes.

The AVRO trial decisively demonstrated the superiority of vernakalant in achieving rapid cardioversion. The primary endpoint was met by 51.7% of patients in the vernakalant group, compared to just 5.2% in the amiodarone group (p<0.0001).[9] This result solidified vernakalant's primary clinical advantage: its ability to restore sinus rhythm far more quickly than amiodarone, a critical factor in the acute management of symptomatic AF.

The consistency of the ~51% conversion rate across the ACT I, ACT III, and AVRO trials is striking. This suggests a potential pharmacological ceiling effect, where approximately half of the selected patient population with recent-onset AF are "responders" whose arrhythmia is susceptible to termination by vernakalant's mechanism. The other half appear to be resistant within the 90-minute timeframe. This predictability has important clinical implications, as it allows clinicians to anticipate a roughly 50% success rate for rapid conversion while necessitating a clear contingency plan, such as electrical cardioversion, for the expected non-responders.

Efficacy in Post-Cardiac Surgery Atrial Fibrillation

Postoperative AF is a common complication of cardiac surgery, and vernakalant is specifically indicated for this patient population (AF duration ≤3 days).[1] Its efficacy was evaluated in a prospective, randomized, double-blind, placebo-controlled trial (NCT00125320) in patients who developed AF after coronary artery bypass graft and/or valvular surgery.[35] In this study, vernakalant was significantly more effective than placebo, converting 47% of patients to sinus rhythm within 90 minutes, compared to 14% in the placebo group (

p<0.001).[35] The median time to conversion was 12 minutes. Notably, the trial found that vernakalant was not effective for the conversion of postoperative atrial flutter.[35]

Meta-Analyses and Other Investigations

Subsequent meta-analyses have reinforced the findings from individual trials. A 2022 systematic review confirmed that vernakalant was significantly superior to placebo (Odds Ratio for conversion: 14.94), flecainide (OR: 2.44), and amiodarone (OR: 17.42) for converting recent-onset AF within 90 minutes.[36] Real-world data from post-marketing registries in Europe have reported high conversion rates, ranging from 65% to over 80% in some studies, confirming its effectiveness in clinical practice and its role in facilitating early hospital discharge.[8]

An oral formulation of vernakalant was also investigated for the long-term prevention of AF recurrence. A Phase II study (NCT00526136) found that a 500 mg twice-daily dose significantly delayed the time to AF recurrence compared to placebo and was well-tolerated.[28] However, further clinical development of the oral formulation was ultimately halted.[6] The clinical development program also saw the termination of a Phase 3b study (ACT V, NCT00989001) following a single serious adverse event of cardiogenic shock, which underscored the safety concerns that have shadowed the drug.[2]

Table 5.1: Summary of Efficacy Outcomes from Pivotal Intravenous Vernakalant Trials

Trial Name / IdentifierComparatorPatient Population (AF Duration)N (Vernakalant)Conversion Rate (Vernakalant)Conversion Rate (Comparator)Median Time to ConversionSource(s)
ACT IPlaceboShort duration (3h to 7d)~14551.7%4.0%11 minutes7
ACT IIIPlaceboShort duration (3h to 7d)-51.2%3.6%8 minutes7
AVRO (NCT00668759)AmiodaroneRecent onset (3h to 48h)11651.7%5.2%11 minutes9
Post-Op (NCT00125320)PlaceboPost-cardiac surgery (3h to 72h)10047.0%14.0%12 minutes35

Safety Profile, Tolerability, and Risk Management

The clinical utility of vernakalant is sharply defined by its safety profile. While it offers rapid efficacy, its use is associated with a distinct set of adverse events, particularly cardiovascular, that necessitate careful patient selection and rigorous monitoring. The balance between its benefits and risks is the central issue that has shaped its clinical application and divergent regulatory history.

Adverse Drug Reaction Overview

The adverse events associated with vernakalant can be categorized into common, transient effects and less common but more serious cardiovascular events.

  • Common and Transient Events: The most frequently reported side effects are often mild and resolve shortly after the infusion is completed. These include dysgeusia (a metallic or altered sense of taste), sneezing, and paresthesia (a tingling or numbness sensation).[13] These symptoms are believed to result from the drug's effect on sodium channels in the central nervous system.[2] Other commonly reported non-cardiac effects include dizziness, headache, nausea, and cough.[2]
  • Serious Cardiovascular Events: The adverse events of greatest clinical concern are hemodynamic and electrophysiological.
  • Hypotension: A sudden drop in blood pressure is a primary and well-documented risk of vernakalant administration.[2] Hypotensive events typically occur early, either during the infusion or shortly thereafter, and are reported more frequently in patients with a history of congestive heart failure.[2] The potential for severe, life-threatening hypotension was a key factor in the FDA's decision not to approve the drug.[12]
  • Bradycardia: Vernakalant can cause clinically significant bradycardia, including sinus arrest, particularly after conversion to sinus rhythm.[2] While often responsive to discontinuation of the drug or administration of atropine, some cases have required temporary electrical pacing.[11]
  • Ventricular Arrhythmias: Although vernakalant's minimal effect on the IKr​ current gives it a low intrinsic risk for Torsade de Pointes, other ventricular arrhythmias can occur.[6] Non-sustained ventricular tachycardia has been observed more frequently in vernakalant-treated patients with a history of heart failure (7.3%) compared to placebo (1.6%) in the same population.[7] A fatal case involving ventricular arrhythmia and cardiogenic shock in a patient without apparent structural heart disease was a pivotal event in the drug's regulatory review in the U.S..[12]
  • Atrial Flutter: An increased incidence of atrial flutter, sometimes with rapid 1:1 atrioventricular conduction, has been reported following vernakalant administration.[1]

Contraindications

Due to the risk of severe adverse events, vernakalant is strictly contraindicated in several patient populations where the risk of hemodynamic collapse or proarrhythmia is unacceptably high.

  • Hemodynamic Instability or Severe Structural Disease: Patients with severe aortic stenosis, systolic blood pressure below 100 mmHg, or advanced heart failure (New York Heart Association Class III or IV) must not receive vernakalant.[2]
  • Recent Acute Coronary Syndrome (ACS): Administration is contraindicated in patients who have experienced an ACS, including myocardial infarction, within the preceding 30 days.[2]
  • Baseline Electrophysiological Abnormalities: Patients with a prolonged QT interval at baseline (uncorrected >440 msec), severe bradycardia, sinus node dysfunction, or second- or third-degree atrioventricular block (in the absence of a functioning pacemaker) are contraindicated.[2]
  • Concurrent Antiarrhythmic Use: The use of other intravenous Class I or Class III antiarrhythmic drugs within 4 hours prior to, or 4 hours following, vernakalant administration is contraindicated.[2]

This extensive list of contraindications creates a significant treatment paradox: many of the patients who are most symptomatic and in greatest need of rapid cardioversion due to underlying cardiac disease are the very patients for whom the drug is deemed too dangerous. This effectively limits its use to a healthier, more stable subset of the AF population, who may have other viable, albeit potentially slower, treatment options.

Warnings and Precautions for Use

To mitigate the known risks, the use of vernakalant is governed by stringent warnings and precautions.

  • Monitored Clinical Setting: Vernakalant must only be administered by a qualified healthcare professional in a setting appropriate for cardiac monitoring and cardioversion, with resuscitation equipment readily available.[2]
  • Continuous Monitoring: Continuous electrocardiographic (ECG) and vital sign (blood pressure, heart rate) monitoring is mandatory during the infusion and for at least two hours after administration begins.[11] The infusion must be stopped immediately if significant hypotension or bradycardia occurs.[26]
  • Careful Patient Selection: In addition to the absolute contraindications, vernakalant is not recommended for patients with a left ventricular ejection fraction of 35% or less, hypertrophic obstructive cardiomyopathy, or advanced hepatic impairment.[2] It should be used with caution in patients with NYHA Class I or II heart failure due to the elevated risk of adverse events.[2]
  • Pre-Infusion Assessment: Prior to administration, patients must be adequately hydrated, and any electrolyte abnormalities, particularly hypokalemia, must be corrected.[2] A pre-infusion checklist is often supplied with the product to guide clinicians through the eligibility criteria.[40]

Significant Drug-Drug Interactions

The potential for drug-drug interactions with vernakalant must be carefully considered, particularly given its use in an acute setting where patients may be on multiple concomitant medications. Interactions can be broadly categorized as pharmacodynamic (additive effects on cardiac function) or pharmacokinetic (alterations in drug metabolism). Of these, the immediate pharmacodynamic interactions are of the most critical clinical concern.

Pharmacodynamic Interactions

These interactions involve the additive effects of vernakalant and other drugs on cardiac electrophysiology and hemodynamics.

  • Other Antiarrhythmic Drugs: The most significant interaction is with other antiarrhythmic agents. Due to the high risk of cumulative proarrhythmic effects, the co-administration of intravenous Class I and Class III antiarrhythmics is contraindicated within a 4-hour window (before or after) of vernakalant infusion.[2] Caution is also advised when transitioning to or from oral antiarrhythmics.[41]
  • QTc-Prolonging Agents: Although vernakalant has a minimal effect on the QT interval, its co-administration with other drugs known to prolong the QTc interval could theoretically increase the risk of ventricular tachyarrhythmias. This includes a wide range of medications such as certain macrolide antibiotics (e.g., clarithromycin, azithromycin), antipsychotics (e.g., amisulpride), antidepressants (e.g., citalopram), and antihistamines (e.g., acrivastine).[1]
  • Agents with Negative Chronotropic or Inotropic Effects: The arrhythmogenic potential of vernakalant may be increased when combined with drugs that slow heart rate or depress myocardial contractility. This includes beta-blockers (e.g., acebutolol, atenolol), certain calcium channel blockers, and cardiac glycosides (e.g., acetyldigitoxin).[1] Co-administration requires heightened vigilance and cardiovascular monitoring.

Pharmacokinetic Interactions

These interactions primarily involve the CYP2D6 metabolic pathway.

  • CYP2D6 Inhibitors: Vernakalant is a substrate of the CYP2D6 enzyme. Co-administration with potent inhibitors of this enzyme can decrease the metabolism of vernakalant, leading to higher plasma concentrations and a prolonged elimination half-life. This could increase the magnitude and duration of its effects, including adverse events. Examples of potent CYP2D6 inhibitors include certain antidepressants (e.g., bupropion, fluoxetine, paroxetine) and other drugs like abiraterone and quinidine.[1]
  • CYP2D6 Inducers: Conversely, co-administration with strong CYP2D6 inducers could potentially increase the metabolism of vernakalant, possibly reducing its efficacy, although this is less well-documented.
  • Vernakalant as a CYP2D6 Inhibitor: Vernakalant itself is a moderate inhibitor of CYP2D6. Therefore, it has the potential to increase the plasma concentrations of other drugs that are substrates of this enzyme, such as the tricyclic antidepressant amitriptyline, the antipsychotic aripiprazole, and the beta-blocker metoprolol.[1] However, given vernakalant's short half-life and acute-use setting, the clinical significance of this interaction for chronically administered drugs is likely limited.

Table 7.1: Clinically Significant Drug Interactions with Vernakalant

Interacting Drug/ClassPotential EffectMechanismManagement RecommendationSource(s)
Class I & III Antiarrhythmics (IV)Increased risk of proarrhythmia, conduction defectsAdditive pharmacodynamic effectsContraindicated within 4 hours before or after Vernakalant2
QTc Prolonging AgentsIncreased risk of Torsade de PointesAdditive pharmacodynamic effect on cardiac repolarizationAvoid co-administration; requires careful ECG monitoring if use is unavoidable1
Potent CYP2D6 InhibitorsIncreased Vernakalant exposure and half-lifePharmacokinetic (inhibition of metabolism)Use with caution; monitor closely for adverse events like hypotension and bradycardia1
Beta-blockers, DigoxinIncreased risk of arrhythmogenic activities, bradycardiaAdditive pharmacodynamic effects (e.g., on AV conduction)Use with caution; monitor heart rate and ECG closely1

Regulatory Status and Clinical Perspective

The regulatory journey of vernakalant is a compelling case study in differing risk-benefit assessments by global health authorities, leading to its approval in many parts of the world but repeated rejection in the United States. This divergence highlights the profound impact of its safety profile on its perceived clinical value.

Divergent Regulatory Outcomes: EMA vs. FDA

  • European Medicines Agency (EMA) and Other Jurisdictions: Vernakalant, under the brand name Brinavess, received marketing authorization valid throughout the European Union on September 1, 2010.[5] The EMA's Committee for Medicinal Products for Human Use (CHMP) concluded that, for the specified indication of rapid cardioversion of recent-onset AF in carefully selected patients, the drug's demonstrated benefits in efficacy and speed of action outweighed its known risks.[5] This approval was contingent upon its use by qualified professionals in a monitored setting with strict adherence to contraindications.[5] Subsequently, the drug has been approved in Canada and over 40 countries worldwide, establishing its role in the therapeutic armamentarium in these regions.[1]
  • U.S. Food and Drug Administration (FDA): In stark contrast, vernakalant has failed to gain approval in the United States. The process has been protracted and marked by persistent safety concerns. In 2007, an FDA advisory committee initially voted in favor of recommending approval.[14] However, in 2008, the FDA declined to grant full approval, instead issuing an "approvable letter" that requested additional information, effectively halting its path to market.[14] The manufacturer later resubmitted the New Drug Application with additional data, including from the post-marketing SPECTRUM registry. Despite this, in a decisive meeting in December 2019, the FDA's Cardiovascular and Renal Drugs Advisory Committee voted overwhelmingly (11 to 2) against recommending approval.[12] The panel's decision was heavily influenced by the drug's safety profile, with members citing the risk of severe hypotension, suppression of left ventricular function, and a widely discussed fatal case of ventricular arrhythmia and cardiogenic shock.[12] The FDA's own medical officers described the safety results from the registry as "not reassuring," concluding that the potential for rare but catastrophic adverse events outweighed the clinical benefit of rapid cardioversion, especially given the availability of alternative treatments.[12]

This divergence likely reflects a fundamental difference in regulatory philosophy. The EMA's decision suggests a willingness to approve a highly effective but risky drug for a niche indication, trusting that strict labeling and professional practice can manage the risk. The FDA's repeated rejections indicate a lower tolerance for such risks, particularly for a non-life-threatening condition, and perhaps a greater concern that in a large and diverse healthcare system like the U.S., the necessary restrictions on use might not be consistently followed in real-world practice.

Place in Therapy and Clinical Perspective

Where it is approved, vernakalant occupies a specific niche in the management of AF. Its primary role is as a first-line agent for the rapid pharmacological cardioversion of hemodynamically stable patients with recent-onset AF who have no significant structural heart disease or other contraindications.[2] Its principal advantage is speed. The ability to restore sinus rhythm in a median time of around 11 minutes offers rapid symptom relief and has been shown to facilitate earlier discharge from the emergency department, potentially avoiding hospital admission and reducing healthcare costs.[9]

However, its utility is constrained by its extensive list of contraindications and the absolute requirement for administration in a fully monitored environment with resuscitation capabilities.[8] The predictable failure rate of approximately 50% also means that clinicians must always be prepared to proceed to alternative strategies, such as electrical cardioversion, in a timely manner.

In conclusion, vernakalant is a potent and innovative antiarrhythmic agent whose unique, atrial-selective mechanism provides a clear efficacy benefit in a specific clinical scenario. Its story underscores the critical balance between efficacy and safety in drug development and regulation. While its rapid action is a significant advantage, the potential for severe hemodynamic adverse events has rightfully limited its application to a narrow population of patients in whom the benefits are judged to clearly outweigh the risks. The contrasting decisions of the EMA and FDA serve as a powerful reminder that the definition of an acceptable risk-benefit profile can vary, ultimately shaping the availability of therapeutic options for patients worldwide.

Works cited

  1. Vernakalant: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06217
  2. Introducing Vernakalant into Clinical Practice - PMC - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6434499/
  3. Vernakalant | C20H31NO4 | CID 9930049 - PubChem, accessed September 13, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Vernakalant
  4. Brinavess (vernakalant) dosing, indications, interactions, adverse effects, and more, accessed September 13, 2025, https://reference.medscape.com/drug/brinavess-vernakalant-4000007
  5. Brinavess | European Medicines Agency (EMA), accessed September 13, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/brinavess
  6. Safety and efficacy of vernakalant for acute cardioversion of atrial fibrillation: an update - PMC - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3639220/
  7. Investigational BRINAVESS™ (vernakalant) for Infusion Recommended for Approval by the CHMP in the European Union (EU) for | Fierce Pharma, accessed September 13, 2025, https://www.fiercepharma.com/pharma/investigational-brinavess%E2%84%A2-vernakalant-for-infusion-recommended-for-approval-by-chmp
  8. Introducing Vernakalant into Clinical Practice | AER Journal, accessed September 13, 2025, https://www.aerjournal.com/articles/introducing-vernakalant-clinical-practice?language_content_entity=en
  9. A Randomized Active-Controlled Study Comparing the Efficacy and ..., accessed September 13, 2025, https://www.jacc.org/doi/10.1016/j.jacc.2010.07.046
  10. A randomized active-controlled study comparing the efficacy and safety of vernakalant to amiodarone in recent-onset atrial fibrillation - PubMed, accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/21232669/
  11. Vernakalant - [Product Monograph Template - Standard], accessed September 13, 2025, https://pdf.hres.ca/dpd_pm/00038441.PDF
  12. FDA panel rejects vernakalant bid for AFib cardioversion indication - MDEdge, accessed September 13, 2025, https://www.mdedge.com/jcomjournal/article/214099/arrhythmias-ep/fda-panel-rejects-vernakalant-bid-afib-cardioversion
  13. Vernakalant hydrochloride for rapid conversion of atrial fibrillation: a phase 3, randomized, placebo-controlled trial - PubMed, accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/18332267/
  14. Vernakalant - Wikipedia, accessed September 13, 2025, https://en.wikipedia.org/wiki/Vernakalant
  15. Efficacy and safety of intravenous vernakalant for the rapid conversion of recent‐onset atrial fibrillation: A meta‐analysis - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6931449/
  16. Vernakalant (RSD1235) | Potassium Channel Inhibitor | MedChemExpress, accessed September 13, 2025, https://www.medchemexpress.com/vernakalant.html
  17. VERNAKALANT HYDROCHLORIDE, accessed September 13, 2025, https://searchusan.ama-assn.org/usan/documentDownload?uri=/unstructured/binary/usan/vernakalant_hcl.pdf
  18. Vernakalant Hydrochloride | C20H32ClNO4 | CID 9930048 - PubChem, accessed September 13, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/9930048
  19. Vernakalant Hydrochloride|Ion channel blocker,investigational antiarrhythmic - APExBIO, accessed September 13, 2025, https://www.apexbt.com/vernakalant-hydrochloride.html
  20. Compound: VERNAKALANT (CHEMBL2111112) - ChEMBL - EMBL-EBI, accessed September 13, 2025, https://www.ebi.ac.uk/chembl/explore/compound/CHEMBL2111112
  21. Vernakalant Hydrochloride | CAS 748810-28-8 | SCBT, accessed September 13, 2025, https://www.scbt.com/p/vernakalant-hydrochloride-748810-28-8
  22. Vernakalant hydrochloride | DrugBank Online, accessed September 13, 2025, https://go.drugbank.com/salts/DBSALT002376
  23. VERNAKALANT - Inxight Drugs, accessed September 13, 2025, https://drugs.ncats.io/drug/9G468C8B13
  24. Vernakalant hydrochloride in the treatment of atrial fibrillation: a review of the latest clinical evidence - Open Access Journals, accessed September 13, 2025, https://www.openaccessjournals.com/articles/vernakalant-hydrochloride-in-the-treatment-of-atrial-fibrillation-a-review-of-the-latest-clinical-evidence.pdf
  25. FDA panel rejects vernakalant bid for AFib cardioversion indication | MDedge, accessed September 13, 2025, https://blogs.the-hospitalist.org/content/fda-panel-rejects-vernakalant-bid-afib-cardioversion-indication
  26. Brinavess 20 mg/ml concentrate for solution for infusion - Summary of Product Characteristics (SmPC) - (emc) | 7249, accessed September 13, 2025, https://www.medicines.org.uk/emc/product/7249/smpc
  27. Suitability, Efficacy, and Safety of Vernakalant for New Onset Atrial ..., accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4036718/
  28. Vernakalant Completed Phase 2 Trials for Atrial Fibrillation Prevention | DrugBank Online, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06217/clinical_trials?conditions=DBCOND0000503&phase=2&purpose=prevention&status=completed
  29. A Randomized, Placebo-Controlled Study of Vernakalant (Oral) for the Prevention of Atrial Fibrillation Recurrence After Cardioversion | Circulation: Arrhythmia and Electrophysiology - AHA Journals, accessed September 13, 2025, https://www.ahajournals.org/doi/10.1161/circep.111.962340
  30. The Vernakalant Story: How Did It Come to Approval in Europe and What is the Delay in the U.S.A? - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4101194/
  31. Pharmacological cardioversion of atrial fibrillation with vernakalant: evidence in support of the ESC Guidelines | EP Europace | Oxford Academic, accessed September 13, 2025, https://academic.oup.com/europace/article/16/2/162/524792
  32. A Phase III Superiority Study of Vernakalant vs Amiodarone in Subjects With Recent Onset Atrial Fibrillation (AVRO) - ClinicalTrials.gov, accessed September 13, 2025, https://www.clinicaltrials.gov/study/NCT00668759
  33. A Randomized Active-Controlled Study Comparing the Efficacy and Safety of Vernakalant to Amiodarone in Recent-Onset Atrial Fibrillation | JACC, accessed September 13, 2025, https://www.jacc.org/doi/abs/10.1016/j.jacc.2010.07.046
  34. A Phase III Superiority Study of Vernakalant vs. Amiodarone in Subjects With Recent Onset Atrial Fibrillation - AVRO - American College of Cardiology, accessed September 13, 2025, https://www.acc.org/latest-in-cardiology/clinical-trials/2011/01/12/13/42/avro
  35. Vernakalant Hydrochloride for the Rapid Conversion of Atrial Fibrillation After Cardiac Surgery | Circulation: Arrhythmia and Electrophysiology - AHA Journals, accessed September 13, 2025, https://www.ahajournals.org/doi/10.1161/circep.109.870204
  36. Abstract 11662: Efficacy And Safety Outcome Of Intravenous Vernakalant For Recent-onset Atrial Fibrillation: A Systematic Review And Meta-analysis, accessed September 13, 2025, https://www.ahajournals.org/doi/10.1161/circ.148.suppl_1.11662
  37. Abstract 11662: Efficacy And Safety Outcome Of Intravenous Vernakalant For Recent-onset Atrial Fibrillation: A Systematic Review And Meta-analysis | Request PDF - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/publication/376675165_Abstract_11662_Efficacy_And_Safety_Outcome_Of_Intravenous_Vernakalant_For_Recent-onset_Atrial_Fibrillation_A_Systematic_Review_And_Meta-analysis
  38. Vernakalant for Cardioversion of Recent-Onset Atrial Fibrillation in the Emergency Department: The SPECTRUM Study - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9808723/
  39. Vernakalant Terminated Phase 3 Trials for Atrial Fibrillation Treatment | DrugBank Online, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06217/clinical_trials?conditions=DBCOND0000503&phase=3&purpose=treatment&status=terminated
  40. APPROPRIATE USE OF BRINAVESS® - EMed, accessed September 13, 2025, https://emed.ie/_docs/Pharm-Brinavess_HCP_Card_Ireland_20150701.pdf
  41. BRINAVESS, INN-vernakalant hydrochloride, accessed September 13, 2025, https://ec.europa.eu/health/documents/community-register/2010/2010090184941/anx_84941_en.pdf
  42. Efficacy and Safety of Intravenous Vernakalant in Rapid Cardioversion of Recent Onset Atrial Fibrillation: A Retrospective Single-Centre Study - PubMed, accessed September 13, 2025, https://pubmed.ncbi.nlm.nih.gov/38770450/
  43. Vernakalant (Brinavess) - EMed, accessed September 13, 2025, https://emed.ie/Pharmacology/Vernakalant.php
  44. go.drugbank.com, accessed September 13, 2025, https://go.drugbank.com/drugs/DB06217#:~:text=The%20metabolism%20of%20Vernakalant%20can%20be%20decreased%20when%20combined%20with%20Cisapride.&text=Citalopram-,The%20risk%20or%20severity%20of%20QTc%20prolongation%20can%20be,Vernakalant%20is%20combined%20with%20Citalopram.&text=Clarithromycin-,The%20risk%20or%20severity%20of%20QTc%20prolongation%20can%20be,Vernakalant%20is%20combined%20with%20Clarithromycin.
  45. Brinavess (vernakalant): What is it and is it FDA approved? - Drugs.com, accessed September 13, 2025, https://www.drugs.com/history/brinavess.html

Published at: September 13, 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.