Course

Anti-Arrhythmics 

Course Highlights


  • In this Anti-Arrhythmics course, we will learn about antiarrhythmic drugs according to the Vaughan-Williams classification system.
  • You’ll also learn the mechanisms by which these drugs modify the cardiac action potential to prevent or treat arrhythmias.
  • You’ll leave this course with a broader understanding of the clinical indications for the use of antiarrhythmic drugs, including the management of atrial fibrillation, supraventricular tachycardia, and ventricular arrhythmias.

About

Pharmacology Contact Hours Awarded: 2

Course By:
R.E. Hengsterman

MSN, RN

Begin Now

Read Course  |  Complete Survey  |  Claim Credit

Read and Learn

The following course content

Introduction   

Cardiac arrhythmias continue to present significant clinical challenges and remain a cause common of death and disability [5]. Arrhythmias encompass a wide array of heart rate and rhythm disturbances; they are classified into broad terms as bradyarrhythmia’s (heart rates below 60 beats per minute) and tachyarrhythmias (heart rates exceeding 100 beats per minute) [10].

The defining feature of cardiac arrhythmias is an irregular heartbeat, or a symptom of abnormal heart rhythm linked to irregular initiation of electrical impulses, or a combination of both factors.

The mechanisms underlying cardiac arrhythmias are complex. The management of these conditions often involves the administration of antiarrhythmic drugs. It is vital that prescribers are aware of the pharmacokinetics of this drug class.

Overview of Cardiac Arrhythmias 

Arrhythmias impact an estimated 17 million individuals across the planet, they rank among the most prevalent forms of heart disease [8]. The clinical manifestations of arrhythmias vary from asymptomatic individuals to sudden cardiac death (SCD), contributing to 10–15% of all mortality cases [10].  

The only pattern considered to be a normal heart rhythm is the normal sinus rhythm. In this state, an electrical impulse originates in the sinoatrial (SA) node and travels through the heart [1]. This delayed impulse occurs in the atrioventricular (AV) node, then it proceeds through the His-Purkinje network, which encompasses the bundle of HIS, the left and right bundle branches, and the Purkinje fibers [1]. These reactions essentially coordinate each beat of the heart. 

Despite the notable limitations associated with existing antiarrhythmic medications, pharmacological intervention remains a fundamental aspect of managing cardiac arrhythmias [5]. Research suggests that arrhythmias occur in 1.5% to 5% of the general population, with atrial fibrillation being the most prevalent form [1, 10].  

Arrhythmia refers to any deviation from the heart's normal rhythm, with the normal sinus rhythm being the baseline for a healthy heart rhythm [1]. The categorization of arrhythmias occurs through various criteria, with the most prevalent method being the heart rate of conduction. This includes bradyarrhythmia’s, where the heart beats slower than 60 beats per minute (bpm), and tachyarrhythmias, where the heart rate exceeds 100 bpm [1].  

Atrial fibrillation (AF) is the most frequent occurring sustained cardiac arrhythmia and linked to heightened morbidity and mortality rates, in addition to increased healthcare costs [2][3].  

Arrhythmias may originate from congenital anomalies (present from birth) or can develop due to irritation or damage to the myocardial tissue, causing disruptions or ‘short circuits’ in the heart's electrical system [13].  

 

Basics of Antiarrhythmic Drugs (AADs) 

Antiarrhythmic drugs (AADs) continue to be fundamental in the management of cardiac arrhythmias and classified based on the cardiac action potential [4]. The action of these drugs works toward the immediate cessation of atrial and ventricular arrhythmias (acute cardioversion) and for the long-term prevention of arrhythmia recurrence to maintain normal sinus rhythm [5].  

A limited number of new AADs have reached the market despite the growing incidence of cardiac arrhythmias [5].  

Two key objectives define the rationale for treating arrhythmias: (1) to mitigate significant clinical symptoms, including weakness, syncope, or the onset or worsening of congestive heart failure caused by an arrhythmia, and (2) to extend the patient's lifespan [8].  

In the initial stages of antiarrhythmic drug (AAD) development, the primary focus was on controlling ventricular arrhythmias [6]. The direction of treatment changed following the adverse outcomes highlighted by the Cardiac Arrhythmia Suppression Trial (CAST) and the Survival with Oral D-Sotalol (SWORD) trial, which demonstrated that patients receiving AADs (encainide or flecainide) fared worse than those on placebo [6].  

The primary criterion of antiarrhythmic drugs is safety. In the last decade antiarrhythmic drugs have been subject to intense reevaluation, prompted by the outcomes of large-scale human research studies that highlighted various risks and limitations associated with pharmacological treatments for arrhythmias [7].  

In the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study, the trial demonstrated that although achieving rhythm control was associated with improved survival the adverse effects negated survival benefits [8].  

Quiz Questions

Self Quiz

Ask yourself...

  1. How would you describe the range from asymptomatic cases to life-threatening conditions? 
  2. How can healthcare professionals balance the immediate need for rhythm control with the long-term goal of minimizing adverse effects and mortality? 

Definition 

Antiarrhythmic agents, often referred to as cardiac dysrhythmia medications, constitute a category of pharmaceuticals designed to moderate the heart’s electrical impulse and mitigate rapid heart rhythms; these rhythms include atrial fibrillation (AF), supraventricular tachycardia (SVT), and ventricular tachycardia (VT) [11].  

This class of drugs influence cardiac ionic channels or receptors, modifying the cardiac action potential, or its creation and transmission [11]. These alterations affect the activation spread or repolarization pattern, suppressing cardiac arrhythmias [12].  

Common symptoms of arrhythmias include heart fluttering, abnormal or rapid heart rhythms, lightheadedness, fainting spells, chest pain, and breathlessness. Individuals may also experience heart palpitations, dizziness or feeling faint, chest pain or discomfort, weakness, and fatigue [12]. 

 

 

[10] 

Quiz Questions

Self Quiz

Ask yourself...

  1. How do antiarrhythmic agents contribute to the suppression of arrhythmias and the alleviation of associated symptoms such as heart fluttering, lightheadedness, and chest pain? 
  2. Do you have experience administering antiarrhythmic medications to patients? 

Classification of Antiarrhythmic Medications 

The categorization of arrhythmias is based on the location within the conduction pathway where they originate.  

There are two main groups:  

  • Supraventricular - originating from the atria or the atrioventricular (AV) node. 
  • Ventricular - occur distal to the AV node.  

[23] 

 

Prior to 2018, the categorization of antiarrhythmic drugs was based on the Vaughan-Williams (VW) classification system, which organized these medications by their principal mechanism of action [4, 14]. The initial Vaughan-Williams classification had limitations. When introduced in the 1970s, the range of antiarrhythmic drugs focused on altering the function of Na+, K+, and Ca2+ channels, as well as targeting intracellular processes governed by adrenergic activity [15]. 

Today, there is a broader range of advanced antiarrhythmics, many possessing overlapping interactions with drugs in other classes. For example, amiodarone, categorized under Class III (Potassium channels blockers), also exhibits sodium and calcium-channel blocking properties (Class IV).

Quiz Questions

Self Quiz

Ask yourself...

  1. How do the limitations of the initial Vaughan-Williams classification system highlight the importance of continuous research and development in the field of cardiac pharmacology? 
  2. Can you think of an example of a drug that contains overlapping drug actions? 
Class 0: HCN Channel Blockers 

 

Ivabradine 

Ivabradine is designed to lower heart rate. It is used in the management of stable angina pectoris and chronic heart failure with heart rate ≥70 bpm across various clinical scenarios, including those with either preserved or compromised left ventricular (LV) function.  

Ivabradine is effective by decreasing heart rate while preserving myocardial contractility and coronary vasomotor responsiveness, thereby reducing oxygen consumption, and extending diastolic duration [16] [17]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How does Ivabradine's mechanism of action contribute to its therapeutic benefits in patients with stable angina pectoris and chronic heart failure? 
Class I: Voltage-gated Na+ Channel Blockers 

Class IA 

Class IA medications block fast sodium channels and include agents such as quinidine, procainamide, and disopyramide [11]. Quinidine, disopyramide, and procainamide are used in the management of supraventricular tachyarrhythmias, recurrent atrial fibrillation, ventricular tachycardia, ventricular fibrillation, Brugada syndrome, and Short QT Syndrome (SQTS) [4].  

These drugs are associated with the highest risk of proarrhythmic among sodium channel blockers due to their capacity to prolong the QTc interval, which restricts their use because of their proarrhythmic potential [11].  

For patients with Brugada syndrome, Quinidine is an alternative to implantable cardioverter-defibrillator (ICD) placement [66].  

This class has also shown utility in individuals with short QT syndrome experiencing recurrent ventricular arrhythmias (VAs), reducing the frequency of ICD shocks in these patients [66].  

Disopyramide is employed in cases of hypertrophic obstructive cardiomyopathy (HOCM), when combined with a beta-blocker or verapamil to alleviate symptoms like angina or dyspnea in patients unresponsive to beta-blockers or verapamil alone [67].  

Procainamide is useful for exposing and diagnosing Brugada syndrome in individuals suspected of the condition but without a confirmed diagnosis [68]. Procainamide has shown to reestablish sinus rhythm in patients with Wolff-Parkinson-White (WPW) syndrome who experience atrial fibrillation (AF) without hemodynamic instability, which is marked by a wide QRS complex or a rapid pre-excited ventricular response [69]. Procainamide may also assist in terminating ventricular tachycardia and other arrhythmias [69]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How might the proarrhythmic potential of these drugs and their ability to prolong the QTc interval influence the decision-making process in selecting an appropriate treatment strategy for patients with conditions like Brugada syndrome, Short QT Syndrome (SQTS), or Wolff-Parkinson-White (WPW) syndrome? 

Class IB

Lidocaine and Mexiletine treat ventricular tachycardia and ventricular fibrillation after a myocardial infarction by inducing a mild blockade of sodium channels [4]. These are not effective for treating atrial arrhythmias [4].  

In the context of long QT syndrome, mexiletine is capable of reducing the QTc interval and has been employed to decrease the incidence of recurrent arrhythmias and the need for interventions by implantable cardioverter-defibrillators (ICDs) [70].  

The effectiveness of Lidocaine diminishes in instances of hypokalemia, necessitating the correction of potassium levels [80].

Quiz Questions

Self Quiz

Ask yourself...

  1. How does the mechanism of sodium channel blockade by Lidocaine and Mexiletine contribute to their differential effectiveness in treating ventricular? 
  2. What implications does this have for their use in managing long QT syndrome and conditions of electrolyte imbalance such as hypokalemia? 

Top of Form 

 

Class IC 

Research recommends Encainide (Enkaid), flecainide (Tambocor), and propafenone (Rythmol SR) for managing supraventricular and ventricular tachyarrhythmias that do not respond to standard treatments, in the absence of underlying structural heart disease. [72].  

Propafenone increases the effects of cyclosporin, desipramine, and theophylline [81]. 

Class IC is used to treat premature ventricular contractions and catecholaminergic polymorphic ventricular tachycardia [18]. These drugs block sodium channels without altering the QT interval and suit ongoing management in individuals with symptomatic supraventricular tachycardia (SVT) who have no structural or ischemic heart disease, and either are unsuitable for or opt against catheter ablation. [11].  

In addition, these agents are effective for the pharmacological cardioversion of atrial fibrillation (AF). The “pill in the pocket” strategy involves patients with paroxysmal AF carrying a loading dose of medication to take at the onset of an AF episode. This approach aims for chemical cardioversion to restore normal rhythm, rather than adhering to a regular maintenance dose regime [71].  

The Cardiac Arrhythmia Suppression Trials (CAST I and II) showed that patients with a history of myocardial infarction, treated with class IC agents (flecainide, encainide, moricizine) to reduce premature ventricular contractions (PVCs), faced a higher mortality risk compared to those receiving placebo [72]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How does the balance between the therapeutic benefits in the absence of structural heart disease and the increased mortality risk with a history of myocardial infarction guide clinical decision-making? 
  2. How would you describe the 'pill in the pocket' strategy versus traditional maintenance dosing? 

Class ID

Ranolazine presents a potential therapeutic option for managing tachyarrhythmias and ventricular tachycardia [19]. 

 

 

 

Class II: Autonomic Inhibitors/Activators 

The literature recommends beta-blockers (BB) for managing the heart rate in individuals with paroxysmal, persistent, or permanent atrial fibrillation (AF) and atrial flutter [73]. Beta-blockers (BB) are also beneficial for long-term management in patients with symptomatic supraventricular tachycardia (SVT) [73].  

Due to their favorable safety profile and efficacy, healthcare professionals can consider beta-blockers as the first-choice therapy for ventricular arrhythmias [39]. Their use is associated with a reduction in adverse cardiac events in conditions such as long QT syndrome and catecholaminergic polymorphic ventricular tachycardia [74].  

For patients exhibiting symptomatic premature ventricular contractions (PVCs) in the absence of underlying heart disease, beta-blocker therapy can help decrease the frequency of recurrent arrhythmias and alleviate symptoms [75]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How does the mechanism of action of beta-blockers contribute to their effectiveness across a spectrum of arrhythmias? 
  2. What factors influence the decision to prioritize beta-blockers as a first-choice therapy in these conditions, considering their impact on reducing adverse cardiac events in long QT syndrome and catecholaminergic polymorphic ventricular tachycardia? 

IIa: Inhibitors including pindolol, carvedilol, timolol, nadolol (non-selective beta-blockers), and bisoprolol, atenolol, metoprolol, esmolol (selective beta-1 blockers) treat rate control in atrial fibrillation, atrial flutter, and ventricular tachyarrhythmia [4]. 

IIb: Activators: The use of Isoproterenol can manage ventricular escape rhythm in cases of complete AV block before pacemaker implantation [4][20]. 

IIc: Inhibitors: Atropine treats symptomatic sinus bradycardia and conduction block [4][21]. 

IId: Activators: For the management of supraventricular tachyarrhythmias, Carbachol, methacholine, and digoxin [4]. 

IIe:Activators: Adenosine for the cessation of paroxysmal supraventricular tachycardia (PSVT) [22]. 

 

Class III: K+ Channel Blockers/Openers 

Potassium channel blockers decrease potassium efflux out of the cell and prolong the QTc interval [4] Amiodarone displays sympatholytic effects as well as sodium and calcium channel blocking properties, leading to reduced conduction through the AV and sinus nodes [4].  

Amiodarone helps maintain sinus rhythm in patients with atrial fibrillation (AF) and those suffering from left ventricular systolic dysfunction [4].  

Amiodarone also stands as a viable choice for pharmacological cardioversion and can help manage ventricular rate in critical patients without pre-excitation, though it is less effective than non-dihydropyridine calcium channel blockers [4] [49].  

Amiodarone is the preferred antiarrhythmic medication for suppressing ventricular arrhythmias (VA) [4]. Administration of intravenous amiodarone may achieve rhythm stabilization in cases of unstable persistent ventricular arrhythmias (VA) following defibrillation [49]. Administering intravenous amiodarone stabilizes rhythm in cases of unstable persistent ventricular arrhythmias (VA) following defibrillation.  

In addition, amiodarone controls ventricular arrhythmias (VA) in patients with ischemic heart disease who are also receiving beta-blocker treatment [4].  

Observations show that Dronedarone reduces hospital admissions for atrial fibrillation (AF) in individuals with a history of non-permanent AF who are in sinus rhythm. However, for patients with permanent AF that cannot convert back to normal sinus rhythm, it is contraindicated due to FDA reviews indicating a significant increase in the risk of cardiovascular death, stroke, and heart failure in these cases [76]. 

Dofetilide is employed in the treatment of atrial arrhythmias and for the acute pharmacological cardioversion of atrial fibrillation or flutter [77].  

Sotalol, combining class II beta-blocker properties and class III potassium channel blocker effects, manages both ventricular and supraventricular arrhythmias. [78].  

Ibutilide (Corvert) targets the treatment of atrial fibrillation or flutter, underlining its specialized use in managing these specific arrhythmias [79]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How do these drug mechanisms contribute to its efficacy in managing both atrial and ventricular arrhythmias 
  2. What considerations should be made when choosing Amiodarone for patients with left ventricular systolic dysfunction or those undergoing pharmacological cardioversion? 
  3. How should clinicians navigate the decision-making process for employing Dronedarone in treating AF, considering the patient's AF status and the potential benefits and risks? 

IIIA: Voltage-dependent K+ channels 

Amiodarone and dronedarone are notable for their role as non-selective potassium (K+) channel blockers, which are crucial in the treatment of unstable ventricular tachycardia and life-threatening recurrent ventricular fibrillation [4]. 

 

Kv11.1 (rapid K+ current) blockers: Dofetilide, almokalant, ibutilide, sematilide, and sotalol manage ventricular tachycardia in patients without prior myocardial infarction or underlying structural heart disease. They also treat Wolff-Parkinson-White (WPW) syndrome when associated with atrial fibrillation [4]. 

Kv1.5 (ultra-rapid K+ current) blockers: Vernakalant serves to convert recent onset atrial fibrillation in patients without structural or ischemic heart disease. It is important to note that the FDA does not approve this specific use of vernakalant [24]. 

IIIb: Metabolically dependent K+ channels blockers: Nicorandil and pinacidil are employed as second-line treatments for stable angina [25]. 

 

Class IV: Ca2+ handling modulators 

Non-dihydropyridine calcium channel blockers, such as diltiazem and verapamil, reduce conduction speed and decelerate signal transmission through the AV node [82]. These medications are effective for controlling the ventricular rate in both acute and chronic cases of atrial fibrillation (AF) and atrial flutter [82].  

In the acute management of stable patients with supraventricular tachycardia (SVT), including focal and multifocal atrial tachycardias, diltiazem and verapamil serve as viable treatment options [4] [39]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. Given the mechanism by which non-dihydropyridine calcium channel blockers like diltiazem and verapamil slow conduction and decelerate signal transmission through the AV node, why are these medications suited for controlling the ventricular rate in atrial fibrillation and atrial flutter? 
  2. How does this mechanism influence their effectiveness in the acute management of various forms of supraventricular tachycardia? 

IVa: Bepridil and falipamil, which block non-selective surface membrane calcium (Ca2+) channels, may manage supraventricular tachyarrhythmias [4]. Verapamil and diltiazem, which block surface membrane L-type calcium (Ca2+) channels, treat supraventricular arrhythmias, and control the rate of atrial fibrillation [4]. 

IVb:Propafenone and flecainide serve as intracellular calcium channel blockers utilized in addressing catecholaminergic polymorphic ventricular tachycardia (CPVT) [4]. 

 

Class V Mechanosensitive channel blockers 

Inhibitors: N-(p-amylcinnamoyl) Anthranilic Acid: Under Research and Not Approved by the FDA [26].  

 

Class VI: Gap junction channel blockers 

Inhibitors: carbenoxolone (under investigation- not FDA approved) [27]. 

 

Class VII: Upstream target modulators 

Omega-3 fatty acids: Eicosapentaenoic Acid and Docosahexaenoic Acid: Reduction in Cardiac Death Risk Post-Myocardial Infarction [28]. 

Statins:  Potential for use in atrial fibrillation 

ACE inhibitors: Captopril, Enalapril, Ramipril, Lisinopril (ACE Inhibitors), and ARBs (Losartan, Telmisartan): Potential Use in Atrial Fibrillation Associated with Heart Failure [29]. 

 

Clinical Prescribing Criteria  

Antiarrhythmic medications are pivotal in managing symptoms and safeguarding against the decline of cardiac function caused by conditions such as tachycardia, irregular rhythms, or desynchrony [4]. The primary objective is to reestablish normal cardiac rhythm and conduction, averting the onset of more severe and fatal arrhythmias. 

Antiarrhythmics have a narrow therapeutic index, indicating a minimal margin between the effective dosage and the onset of toxicity [4]. There is a tenuous balance between suboptimal treatment and the risk of toxic or proarrhythmic effects, underscoring the importance of precise dosing and monitoring. Clinical attention focuses on the patient's clinical status, underlying structural and functional conditions, and the mechanisms of arrhythmia at both cellular and molecular levels.  

The use of antiarrhythmic drugs in therapy seeks to alter conduction velocity, by either slowing down or speeding it up, modify the excitability of cardiac cells via changes in the length of the effective refractory period, and suppress unusual spontaneous activity [14]. 

Numerous variables influence the effectiveness of antiarrhythmic drugs, such as race, sex, genetics, environmental temperature, drug interactions, precipitating factors, changes in neurohormones, the disease's present condition and severity, and disease-driven structural changes in the body [30]. The complexity increases with some antiarrhythmic drugs (AADs) displaying diverse electrophysiological and pharmacological effects that depend on the administration route, plasma concentration, and the existence of active metabolites. 

A variety of factors can influence the efficacy of medications, including racial background, gender, genetic makeup, ambient temperature, interactions between different drugs, initiating triggers, neurohormonal fluctuations, the current state and intensity of the disease, and alterations in the body's structure caused by the disease itself [31].  

Complicating the pharmacology, certain antiarrhythmic drugs (AADs) exhibit a wide range of electrophysiological and pharmacological actions, which can vary based on the method of administration, concentration levels in the plasma, and the presence of active metabolites [4] [5]. 

 

 

Quiz Questions

Self Quiz

Ask yourself...

  1. Considering the narrow therapeutic index of antiarrhythmic drugs, how does precise dosing and monitoring contribute to optimizing treatment outcomes while minimizing the risk of adverse effects? 
  2. What role do patient-specific factors play in this process? 
  3. Given the complex interplay of factors such as race, sex, genetics, and environmental conditions on the effectiveness of antiarrhythmic drugs, how should clinicians integrate this knowledge into personalized treatment plans? 
  4. With some antiarrhythmic drugs displaying varied electrophysiological and pharmacological effects based on administration route, plasma concentration, and the presence of active metabolites, how do these variables complicate the management of arrhythmias? 

The Cardiac Electrical Cycle (Electrical Cascade) 

The cardiac action potential represents the sequence of ion exchanges that result in the successive depolarization and repolarization of the cardiac myocyte, culminating in muscle contraction [4]. During its resting phase, a cardiac myocyte maintains a baseline resting membrane potential ranging from negative 80 to negative 90 millivolts [32].  

Antiarrhythmic drugs slow down ion movement during various stages of the cardiac action potential [32]. 

 

  • Phase 0:The “depolarization” phase of the action potential occurs due to the influx of sodium ions (Na+) into the cell, following an electrochemical gradient, leading to a membrane potential of around positive 30 millivolts [33]. 
  • Phase 1: “The notch,” or the early repolarization phase of the action potential, features potassium (K+) ions flowing out. [4] [33]. 
  • Phase 2:“The plateau” phase occurs when the inward movement of calcium ions (Ca2+) balances the outward movement of potassium (K+) ions [4] [34]. 
  • Phase 3:“The repolarization” phase of the action potential occurs through the efflux of potassium (K+) ions along their electrochemical gradient out of the cell. This movement removes the positive charge of the K+ ion from the cell, reinstating the cardiac myocyte's negative potential [33][34]. 
  • Phase 4:Reactivation of the Na/K-ATPase pump, which re-establishes the resting membrane potential in the cardiac myocyte [4][35]. 
Quiz Questions

Self Quiz

Ask yourself...

  1. How do antiarrhythmic drugs alter the ion movement during the different phases of the cardiac action potential to correct arrhythmias? 
  2. What are the potential consequences of these modifications on the overall function of the cardiac myocyte and the heart's rhythm? 

Pharmacokinetics of Anti-Arrhythmic Medications 

Pharmacokinetics involves the study of drug absorption, distribution, metabolism, and excretion. All antiarrhythmic drugs affect the conductance of membranes and ions, modifying cardiac action potential dynamics either via direct or indirect action.  

For example, some medications inhibit fast sodium channels, essential for controlling the rate of membrane depolarization (phase 0) during an action potential [33]. Electrical conduction velocity links to membrane depolarization and blocking sodium channels slows this velocity down [32][33]. Slowing conduction velocity is advantageous for eradicating tachyarrhythmias resulting from reentry circuits [36].  

Various antiarrhythmic drug classes affect the duration of action potentials and the effective refractory period [37]. Extending the effective refractory period often eradicates reentry tachyarrhythmias [38]. This effect occurs by inhibiting potassium channels and postponing the repolarization phase (phase 3) of action potentials [38].  

Medications that inhibit the slow inward calcium channels aim to diminish pacemaker activity by decelerating the depolarizing pacemaker potential’s rate of rise (phase 4 depolarization) [14].  

These drugs also decrease the speed of electrical signal transmission through the atrioventricular (AV) node [14][38]. Similar to sinoatrial (SA) node cells, AV nodal cells rely on the influx of calcium ions for depolarization [34]. Due to the potential for sympathetic nervous system activity to cause arrhythmias, beta1-adrenoceptor blockers are employed to diminish the sympathetic impact on the heart [39]. These beta-adrenoceptors connect to ion channels through specific signal transduction pathways, indicating that beta-blockers modify ion conductance across the membrane, influencing calcium and potassium conductance [39].  

In instances of AV block, doctors sometimes use drugs like atropine, a muscarinic receptor antagonist, to counteract vagal effects [40]. AV block can emerge as an adverse effect of beta-blocker medication, and stopping the beta-blocker in such cases may return AV conduction to normal [41].  

An increased ventricular rate can be a consequence of atrial flutter or fibrillation [42].  
In response, medications that slow down conduction through the atrioventricular (AV) node regulate the ventricular rate. Calcium channel blockers and beta-blockers are particularly effective for this purpose [4].  

Digoxin proves advantageous for patients with systolic heart failure, also referred to as heart failure with reduced ejection fraction (HFrEF), characterized by an ejection fraction of less than 40% [43]. Nonetheless, it does not contribute to a reduction in mortality [43]. When standard treatments do not achieve heart rate objectives in atrial fibrillation or atrial flutter, clinicians can deploy Digoxin for heart rate management [44].  

Administration of digoxin is contraindicated in instances of pre-excitation due to accessory pathways since it promotes AV blockade and could precipitate ventricular tachyarrhythmias [44]. In conditions of elevated sympathetic activity, digoxin is ineffective, and beta-blockers are the preferred treatment option [44].  

Healthcare providers favor oral drug formulations for their improved patient compliance, ease of use, and scalability, which offer economic advantages [45]. However, the oral bioavailability of drugs can vary, influenced by differences in physicochemical characteristics and metabolic activities that impact pharmacokinetics [46]. Challenges such as intestinal metabolism, efflux mechanisms in the gastrointestinal tract, and the hepatic first-pass effect hinder the bioavailability of drugs administered orally [46] [47].  

The first-pass effect describes a pharmacokinetic process in which a drug undergoes metabolism at a specific site in the body before reaching the systemic circulation or its intended site of action, which reduces the concentration of the active drug available [47]. The liver, a primary location for drug metabolism, has a direct link to the first-pass effect. This effect can also occur in other active metabolic areas of the body, such as the lungs, blood vessels, gastrointestinal tract, and various tissues [47]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How do the pharmacokinetic processes of absorption, distribution, metabolism, and excretion influence the clinical efficacy and safety of antiarrhythmic drugs? 
  2. What strategies can healthcare providers employ to mitigate potential adverse effects, including those related to the first-pass effect? 
  3. Given the varied mechanisms by which antiarrhythmic drugs modify cardiac action potentials and conduction velocities to treat arrhythmias, how do these mechanisms align with the selection of specific antiarrhythmic medications for conditions such as atrial flutter, atrial fibrillation, and AV block, considering both the intended therapeutic outcomes and the potential for adverse effects? 
Absorption 

Antiarrhythmic medications exhibit quick absorption, but the pronounced first-pass effect often reduces their bioavailability [47]. They achieve peak plasma concentrations within 1–3 hours, except for digoxin and dronedarone, which reach their peak in 3–6 hours, and amiodarone, which takes 6–8 hours to peak [11]. In elderly individuals and patients with liver dysfunction, the oral bioavailability of medications tends to be higher [11].  

Intestinal bacteria transform digoxin into inactive compounds; antibiotics such as tetracycline and erythromycin eliminate these bacteria, leading to elevated levels of digoxin in the bloodstream [44]. The antiarrhythmic effect of digoxin starts within 2–5 minutes after its intravenous administration [44]. 

The absorption of drugs through the gastrointestinal tract is crucial for their bioavailability, with meals playing a role that can either enhance or impede this process [46]. For instance, a high-fat meal can increase the oral absorption of dronedarone by fourfold [11]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How do factors such as age, liver function, intestinal flora, and dietary habits influence the therapeutic levels and efficacy of these drugs in the bloodstream? 
Distribution 

With the exception of sotalol, antiarrhythmic drugs (AADs) exhibit some degree of binding to plasma proteins [11]. Amiodarone, digoxin, flecainide, and propafenone build up in the heart at concentrations higher than those in plasma and dialysis cannot remove them [11].  
The concurrent administration of flecainide and amiodarone increases flecainide plasma concentrations by 50% [48].  

Disopyramide, mexiletine, sotalol, and verapamil can cross the placenta and appear in breast milk. High concentrations of Procainamide also occur in breast milk and eliminated by newborns [11].  

Oral administration of amiodarone reaches steady-state plasma concentrations after an extended period, except when administered in substantial loading doses; delivering it via intravenous form also delays its maximal effect [49]. This delay is indicative of its distribution across multiple compartments, including the intravascular compartment, which a standard loading dose saturates, a peripheral compartment encompassing various tissues, and a deep compartment represented by adipose tissue, serving as a reservoir for the drug [49].  

Amiodarone is known for its distinctive side effects, with a 15% prevalence rate in the first year of use, which can escalate to up to 50% with prolonged treatment [87]. Side effects include pulmonary fibrosis, thyroid dysfunction, photosensitivity, blue-grey skin discoloration, corneal microdeposits, peripheral neuropathy, and elevated liver enzymes [87]. Providers must weigh the potential benefits of amiodarone against its long-term risks [94]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How does the differential binding of antiarrhythmic drugs to plasma proteins and their accumulation in various body compartments (heart and adipose tissue) affect their pharmacodynamics and pharmacokinetics? 
Biotransformation 

Antiarrhythmic drugs (AADs) undergo metabolism in the liver through CYP450 isoenzymes, resulting in active metabolites that either block sodium (Na+) channels (such as mexiletine and propafenone), extend action potential duration (APD) [for instance, N-acetylprocainamide (NAPA)], or cause central nervous system (CNS) toxicity (as seen with lidocaine) [11].  

Genetics influence the metabolism of CYP2D6 resulting in higher plasma concentrations and extended half-lives (t½) of metoprolol and propafenone in individuals with poor metabolizing capabilities (6% of Caucasians) compared to those who are rapid metabolizers [50]. In similar fashion, the conversion of procainamide to N-acetylprocainamide (NAPA) varies, with 15–20% metabolized in individuals classified as 'slow-acetylators' and 25–33% in 'fast-acetylators' [11].  

These metabolic phenotypes are determined by genetics. There are no standard tests available to identify a patient's metabolic phenotype prior to treatment initiation, with the exception of measuring the procainamide/NAPA concentration ratio. It is advisable to decrease dosages for poor or slow metabolizers to two-thirds or less of the standard maintenance dose [11] [51].  

Lipophilic beta-blockers, such as bisoprolol, carvedilol, metoprolol, and propranolol, undergo metabolism via CYP2D6, and their bioavailability and half-life (t½) are prolonged in cases of liver dysfunction [11]. The body excretes hydrophilic beta-blockers, including atenolol and sotalol, in their unchanged form through the urine [11].  

Following an intravenous loading dose, lidocaine undergoes metabolism with a half-life (t½) of 1.5–2 hours. In patients with liver impairment or decreased hepatic blood flow — including the elderly, those experiencing cardiogenic shock, heart failure, myocardial infarction, or those taking cimetidine and beta-blockers — lidocaine's plasma levels increase, and its half-life extends [51]. In these cases, one should lower both the loading and maintenance doses.  

Due to its brief half-life, an initial loading dose of lidocaine requires supplementation with a continuous infusion or repeated administrations to achieve and maintain a consistent plasma concentration [53].  

Intravenous Esmolol undergoes rapid hydrolysis in red blood cells, with a half-life of 9 minutes, and achieves complete reversal of beta-blockade 20–30 minutes after drug cessation [11]. Intravenous adenosine acts within 15–30 seconds, with erythrocytes and vascular endothelial cells absorbing and metabolizing it through adenosine deaminase (ADA), leading to a short half-life (t½) of less than 10 seconds [11]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How do genetic variations in CYP450 isoenzymes (CYP2D6) affect the metabolism, efficacy, and safety of antiarrhythmic drugs? 
Elimination 

Antiarrhythmic drugs (AADs) vary in their extent of excretion through urine and feces [11]. The half-life (t½) of these drugs extends in elderly individuals and patients with renal impairment (such as digoxin, disopyramide, dofetilide, flecainide, procainamide, and sotalol) or liver dysfunction (including amiodarone, diltiazem, flecainide, lidocaine, metoprolol, mexiletine, propafenone, propranolol, quinidine, and verapamil) [11]. This prolongation also occurs in congestive heart failure (seen with amiodarone, flecainide, lidocaine, mexiletine, procainamide, and quinidine) or after a myocardial infarction (noted with disopyramide, lidocaine, and mexiletine) [11][54].  

For these patients, it is advisable to lower the dosages and to conduct regular ECG monitoring. Amiodarone is subject to extensive metabolism in the liver, excreted through the bile, and has a prolonged half-life (t½) ranging from 25 to 110 days. This extended half-life accounts for the persistence of its effects for weeks or even months following cessation of the drug [11][55].  

Due to their short half-life (t½), manufacturers dispense certain antiarrhythmic drugs, including beta-blockers, diltiazem, propafenone, and verapamil, in modified-release formulations [11].  

Amiodarone, cimetidine, diltiazem, ketoconazole, procainamide, propranolol, and verapamil elevate plasma concentrations of quinidine [11]. Quinidine acts as a strong inhibitor of CYP2D6 and P-glycoprotein (P-gp), raising the plasma levels of drugs metabolized by this enzyme; it also reduces digoxin clearance, necessitating a 50% reduction in digoxin dosage [11]. Beta-blockers, cimetidine, and halothane cause an increase in plasma concentrations of lidocaine, thereby requiring a reduction in lidocaine dosage [11]. In addition, mexiletine elevates the plasma levels of theophylline, while amiodarone increases the levels of mexiletine [56]. 

Flecainide and propafenone lead to higher plasma concentrations of digoxin and propranolol. Propafenone (Rythmol) raises the plasma levels of digoxin, metoprolol, propranolol, and warfarin [48]. Mexiletine and quinidine amplify the effects of warfarin; thus, it is advisable to decrease the dosage of warfarin and monitor the prothrombin time/international normalized ratio (INR) [11] [48].  

Amiodarone inhibits P-glycoprotein (P-gp) and several cytochrome P450 isoenzymes, such as CYP1A2, CYP2C9, CYP2D6, and CYP3A4, thus increasing the plasma concentrations of drugs metabolized by these pathways or that are substrates of P-gp [94]. Dosage modifications are necessary for medications such as digoxin, flecainide, and warfarin; it is also important to monitor digoxin concentrations and the international normalized ratio (INR) [57][58].  

Cholestyramine may decrease the absorption of amiodarone [59]. Since diltiazem and verapamil inhibit both CYP3A4 and P-glycoprotein (P-gp), adjusting the dosages of drugs metabolized by CYP3A4 or are substrates of P-gp becomes necessary [59]. Furthermore, verapamil has the capacity to suppress the liver's metabolism of lipophilic beta-blockers, resulting in elevated plasma concentrations of these medications [60].  

There is a significant pharmacokinetic interaction between certain antiarrhythmic/rate controlling medications (such as amiodarone, quinidine, dronedarone, verapamil, digoxin, and diltiazem) and non-vitamin K antagonist oral anticoagulants (NOACs) due to competition for P-glycoprotein (P-gp) or inhibition of CYP3A4 (notably by diltiazem, dronedarone, and verapamil) [61] [62].  

Due to these interactions leading to elevated plasma levels of NOACs, experts advise against combining dronedarone with dabigatran and recommend reducing the dose of edoxaban by 50% [62] [63]. Consider reducing the dose of all non-vitamin K antagonist oral anticoagulants (NOACs) when administering amiodarone alongside other P-gp competing substances [64].  

Prescribers recommend reducing the dose of dabigatran (Pradaxa) when used with verapamil. Combining edoxaban with verapamil, especially when other P-gp competitors are present, may also necessitate a dose reduction [65].

Quiz Questions

Self Quiz

Ask yourself...

  1. Considering the extensive metabolism of antiarrhythmic drugs in the liver and their excretion through bile or urine, how do renal and liver dysfunctions affect the pharmacokinetics of these drugs? 
  2. What principles should guide the adjustment of dosages in patients with such conditions to maintain therapeutic efficacy while minimizing toxicity? 
Other Antiarrhythmic Drugs 
  • Adenosine is effective for both diagnosing and halting supraventricular tachycardia (SVT) arising from atrioventricular nodal reentrant tachycardia (AVNRT) or orthodromic atrioventricular reentrant tachycardia (AVRT) [83]. Adenosine serves as a diagnostic aid by revealing underlying atrial flutter or atrial tachycardia (AT) [83]. Adenosine can also terminate focal AT caused by a triggered mechanism and distinguish focal AT from AVNRT and AVRT [83]. Adenosine, a purine nucleoside, results from the breakdown of adenosine triphosphate [86]. Within cardiomyocytes, it interacts with Gi-protein type 1 receptors, facilitating swift potassium efflux and hyperpolarization, while also inhibiting calcium influx [86]. These actions decrease the heart rate and slow down conduction velocity by targeting the AV node. 
  • Digoxin is not a first-line therapy for ventricular rate control in patients with AF, a combination of digoxin and beta-blocker/or non-dihydropyridine calcium channel blockers is a reasonable rate control option in patients with AF and heart failure [84]. 

 

Treatment of Overdose  

In instances of antiarrhythmic drug overdose, medical professionals must ensure the patient has a clear airway, adequate breathing, and support for circulation [4] [87]. Managing cardiac arrest and severe toxicity from poisoning involves the use of specialized interventions, including antidotes and venoarterial extracorporeal membrane oxygenation (VA-ECMO), alongside fundamental and advanced life support techniques [87].  

Symptoms such as nausea, vomiting, neurological manifestations, and lethal arrhythmias characterize Digoxin toxicity [88]. To treat ventricular tachyarrhythmias from digoxin toxicity, clinicians can use lidocaine, and atropine serves as an option for bradyarrhythmia’s. In addition, digoxin-specific antibody fragments prove effective in severe toxicity cases [89].  

Therapeutic and excessive dosages of dofetilide may induce Torsades de Pointes (TdP), which clinicians manage by reducing or stopping the drug's dosage [90]. If the arrhythmia persists, initial treatment involves activated charcoal if ingestion occurred within the last 15 minutes, followed by intravenous magnesium and correction of any electrolyte imbalances [90].  
For persistent arrhythmias, administering isoproterenol/dopamine may serve as a temporary measure until initiating pacing [90]. 

In cases of beta-blocker poisoning, treatments involve administering catecholamines, applying high-dose insulin euglycemic therapy, and using vasopressors, noting glucagon for its positive effects on hemodynamics [91].  

Treating calcium channel blocker (CCB) overdoses involves administering intravenous calcium, dopamine, and norepinephrine. High-dose insulin therapy can reduce mortality in cases of calcium channel blocker poisoning [60]. For severe shock or cardiac arrest resulting from these overdoses, extracorporeal life support is employed [60]. Case reports indicate that clinicians use lipid emulsion therapy to treat overdoses of amiodarone and flecainide [4] [92]. 

Quiz Questions

Self Quiz

Ask yourself...

  1. How do the principles of emergency management in the treatment of severe toxicity from antiarrhythmic drug overdose?  
  2. What factors determine the choice of specific treatments for complications such as lethal arrhythmias, digoxin toxicity, Torsades de Pointes, beta-blocker poisoning, and calcium channel blocker overdoses? 

Conclusion 

Arrhythmias encompass a broad spectrum of heart rate and rhythm disturbances and present significant clinical challenges. Atrial fibrillation (AF) is the most prevalent arrhythmia, associated with increased morbidity, mortality, and healthcare costs [2][3]. Management involves antiarrhythmic drugs (AADs), which play a fundamental role despite their limitations and the potential for adverse effects.  

Antiarrhythmic agents, classified by their primary action mechanism on the cardiac action potential, impact ionic channels or receptors, aiming to suppress arrhythmias [4]. The management objectives for arrhythmias include alleviating significant clinical symptoms and extending life.  

Pharmacokinetic aspects, such as drug absorption, distribution, metabolism, and excretion, play crucial roles in the effectiveness and safety of AADs. Pharmacodynamics involves modifying the cardiac action potential and conduction velocity to prevent or terminate arrhythmias. Factors influencing drug efficacy include genetics, environmental conditions, and the patient’s specific clinical profile. 

Treatment of overdose with antiarrhythmic drugs requires immediate medical intervention, including antidotes and supportive measures like venoarterial extracorporeal membrane oxygenation (VA-ECMO) for severe cases. The management of specific drug toxicities, such as digoxin and beta-blockers, involves targeted therapies and supportive care to mitigate adverse effects and stabilize the patient’s condition. 

References + Disclaimer

  1. Desai, D. S. (2023, June 5). Arrhythmias. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK558923/ 
  2. Benjamin, E. J., Wolf, P. A., D’Agostino, R. B., Silbershatz, H., Kannel, W. B., & Levy, D. (1998). Impact of atrial fibrillation on the risk of death: the Framingham Heart Study. Circulation, 98(10), 946-952.
  3. Atrial fibrillation | Cdc.gov. (2022, October 14). Centers for Disease Control and Prevention. https://www.cdc.gov/heartdisease/atrial_fibrillation.htm
  4. King, G. S. (2023, February 19). Antiarrhythmic medications. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK482322/
  5. Saljic, A., Heijman, J., & Dobrev, D. (2023). Recent Advances in Antiarrhythmic Drug Therapy. Drugs, 83(13), 1147–1160. https://doi.org/10.1007/s40265-023-01923-3
  6. Kowey, P. R., & Robinson, V. M. (2020). The relentless pursuit of new drugs to treat cardiac arrhythmias. Circulation, 141(19), 1507–1509. https://doi.org/10.1161/circulationaha.119.045149
  7. Wright, K. N. (2009). Antiarrhythmic agents. In Elsevier eBooks (pp. 807–810). https://doi.org/10.1016/b978-1-4160-2591-7.10190-0
  8. Geng, M., Lin, A., & Nguyen, T. P. (2020). Revisiting Antiarrhythmic Drug Therapy for Atrial Fibrillation: Reviewing Lessons Learned and Redefining Therapeutic Paradigms. Frontiers in pharmacology, 11, 581837. https://doi.org/10.3389/fphar.2020.581837
  9. Sharma, A.K., Singh, S., Bhat, M. et al. new drug discovery of cardiac anti-arrhythmic drugs: insights in animal models. Sci Rep13, 16420 (2023). https://doi.org/10.1038/s41598-023-41942-4
  10. Heart arrhythmias. (2024). Physiopedia. https://www.physio-pedia.com/Heart_Arrhythmias
  11. Dan, G., Martínez‐Rubio, A., Agewall, S., Boriani, G., Borggrefe, M., Gaïta, F., Van Gelder, I. C., Görenek, B., Kaski, J. C., Kjeldsen, K., Lip, G. Y., Merkely, B., Okumura, K., Piccini, J. P., Potpara, T., Poulsen, B. K., Saba, M., Savelieva, I., Tamargo, J., & Wolpert, C. (2018). Antiarrhythmic drugs–clinical use and clinical decision making: a consensus document from the European Heart Rhythm Association (EHRA) and European Society of Cardiology (ESC) Working Group on Cardiovascular Pharmacology, endorsed by the Heart Rhythm Society (HRS), Asia-Pacific Heart Rhythm Society (APHRS) and International Society of Cardiovascular Pharmacotherapy (ISCP). Europace, 20(5), 731–732an. https://doi.org/10.1093/europace/eux373
  12. Heart arrhythmia – Symptoms and causes – Mayo Clinic. (2023, October 13). Mayo Clinic. https://www.mayoclinic.org/diseases-conditions/heart-arrhythmia/symptoms-causes/syc-20350668
  13. Arrhythmias and congenital defects. (2023, June 15). www.heart.org. https://www.heart.org/en/health-topics/congenital-heart-defects/the-impact-of-congenital-heart-defects/arrhythmias-and-congenital-defects
  14. CV Pharmacology | Vaughan-Williams Classification of Antiarrhythmic Drugs. (2024). https://cvpharmacology.com/antiarrhy/vaughan-williams
  15. Lei, M., Wu, L., Terrar, D. A., & Huang, C. L. (2018). Modernized classification of cardiac antiarrhythmic drugs. Circulation, 138(17), 1879–1896. https://doi.org/10.1161/circulationaha.118.035455
  16. Custodis F, Schirmer SH, Baumhäkel M, et al. Vascular pathophysiology in response to increased heart rate. J Am Coll Cardiol 2010; 56:1973–83. 10.1016/j.jacc.2010.09.014
  17. Kaski, J. C., Gloekler, S., Ferrari, R., Fox, K., Lévy, B., Komajda, M., Vardas, P., & Camici, P. G. (2018). Role of ivabradine in management of stable angina in patients with different clinical profiles. Open Heart, 5(1), e000725. https://doi.org/10.1136/openhrt-2017-000725
  18. Salvage, S. C., Chandrasekharan, K. H., Jeevaratnam, K., Dulhunty, A. F., Thompson, A. J., Jackson, A. P., & Huang, C. L. (2018). Multiple targets for flecainide action: implications for cardiac arrhythmogenesis. British journal of pharmacology, 175(8), 1260–1278. https://doi.org/10.1111/bph.13807
  19. Bazoukis, G., Tse, G., Letsas, K. P., Thomopoulos, C., Naka, K. K., Korantzopoulos, P., Bazoukis, X., Michelongona, P., Papadatos, S. S., Vlachos, K., Liu, T., Efremidis, M., Baranchuk, A., Stavrakis, S., & Tsioufis, C. (2018). Impact of ranolazine on ventricular arrhythmias – A systematic review. Journal of arrhythmia, 34(2), 124–128. https://doi.org/10.1002/joa3.12031
  20. Rahman, M. F. F. (2023, July 24). Atrioventricular dissociation. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK563205/
  21. Perera, R. K., Fischer, T. H., Wagner, M., Dewenter, M., Vettel, C., Bork, N. I., Maier, L. S., Conti, M., Wess, J., El-Armouche, A., Hasenfuß, G., & Nikolaev, V. O. (2017). Atropine augments cardiac contractility by inhibiting cAMP-specific phosphodiesterase type 4. Scientific reports, 7(1), 15222. https://doi.org/10.1038/s41598-017-15632-x
  22. Singh, S. (2023, August 28). Adenosine. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK519049/
  23. Patti, L. (2023, August 7). Supraventricular tachycardia. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK441972/
  24. McIntyre, W. F., Healey, J. S., Bhatnagar, A. K., Wang, P., Gordon, J. A., Baranchuk, A., Deif, B., Whitlock, R. P., & Belley-Côté, É. P. (2019). Vernakalant for cardioversion of recent-onset atrial fibrillation: a systematic review and meta-analysis. Europace: European pacing, arrhythmias, and cardiac electrophysiology: journal of the working groups on cardiac pacing, arrhythmias, and cardiac cellular electrophysiology of the European Society of Cardiology, 21(8), 1159–1166. https://doi.org/10.1093/europace/euz175
  25. Ahmed L. A. (2019). Nicorandil: A drug with ongoing benefits and different mechanisms in various diseased conditions. Indian journal of pharmacology, 51(5), 296–301. https://doi.org/10.4103/ijp.IJP_298_19
  26. PubChem. (2024). N-(p-Amylcinnamoyl) anthranilic acid. PubChem. https://pubchem.ncbi.nlm.nih.gov/compound/N-_p-Amylcinnamoyl_anthranilic-acid
  27. Salcher, S., Spoden, G. A., Hagenbuchner, J., Führer, S., Kaserer, T., Tollinger, M., Huber-Cantonati, P., Gruber, T., Schuster, D., Gust, R., Zwierzina, H., Müller, T., Kiechl‐Kohlendorfer, U., Ausserlechner, M. J., & Obexer, P. (2019). A drug library screen identifies Carbenoxolone as novel FOXO inhibitor that overcomes FOXO3-mediated chemoprotection in high-stage neuroblastoma. Oncogene, 39(5), 1080–1097. https://doi.org/10.1038/s41388-019-1044-7
  28. Le, V. T., Knight, S., Watrous, J. D., Najhawan, M., Dao, K., McCubrey, R. O., Bair, T. L., Horne, B. D., May, H. T., Muhlestein, J. B., Nelson, J. R., Carlquist, J. F., Knowlton, K. U., Jain, M., & Anderson, J. L. (2023). Higher docosahexaenoic acid levels lower the protective impact of eicosapentaenoic acid on long-term major cardiovascular events. Frontiers in cardiovascular medicine, 10, 1229130. https://doi.org/10.3389/fcvm.2023.1229130
  29. Alcocer, L., Bryce, A., Brasil, D. P., Terán, J. L., Cortés, J. E., Quesada, D., & Rodríguez, P. (2023). The pivotal role of Angiotensin-Converting enzyme inhibitors and Angiotensin II receptor blockers in hypertension management and cardiovascular and renal Protection: A Critical appraisal and comparison of international guidelines. American Journal of Cardiovascular Drugs, 23(6), 663–682. https://doi.org/10.1007/s40256-023-00605-5
  30. Kornej, J., Börschel, C. S., Benjamin, E. J., & Schnabel, R. B. (2020). Epidemiology of Atrial fibrillation in the 21st century. Circulation Research, 127(1), 4–20. https://doi.org/10.1161/circresaha.120.316340
  31. O’Brien, T. J., Fenton, K., Sidahmed, A., Barbour, A., & Harralson, A. F. (2021). Race and Drug Toxicity: A Study of Three Cardiovascular Drugs with Strong Pharmacogenetic Recommendations. Journal of Personalized Medicine, 11(11), 1226. https://doi.org/10.3390/jpm11111226
  32. Joukar S. (2021). A comparative review on heart ion channels, action potentials and electrocardiogram in rodents and humans: extrapolation of experimental insights to clinic. Laboratory animal research, 37(1), 25. https://doi.org/10.1186/s42826-021-00102-3
  33. Chrysafides, S. M. (2023, April 10). Physiology, resting potential. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK538338/
  34. Wei, X. (2023, April 17). Physiology, cardiac repolarization dispersion and reserve. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK537194/
  35. Pirahanchi, Y. (2023, March 13). Physiology, Sodium potassium pump. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK537088/
  36. Goyal, A. (2023, July 4). Reentry arrhythmia. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK537089/
  37. An, Z., Yang, G., Liu, X., Zhang, Z., & Liu, G. (2018). New progress in understanding the cellular mechanisms of anti-arrhythmic drugs. Central European Journal of Biology, 13(1), 335–339. https://doi.org/10.1515/biol-2018-0041
  38. Osadchii, O. E. (2017). Effects of Na+ channel blockers on the restitution of refractory period, conduction time, and excitation wavelength in perfused guinea-pig heart. PLOS ONE, 12(2), e0172683. https://doi.org/10.1371/journal.pone.0172683
  39. Wołowiec, Ł., Grześk, G., Osiak, J., Wijata, A., Mędlewska, M., Gaborek, P., Banach, J., Wołowiec, A., & Głowacka, M. (2023). Beta-blockers in cardiac arrhythmias–Clinical pharmacologist’s point of view. Frontiers in Pharmacology, 13. https://doi.org/10.3389/fphar.2022.1043714
  40. Naji, A. (2023, May 8). Muscarinic antagonists. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK557541/
  41. Marcu, D., Adam, C. A., Dorobantu, D. M., Șalaru, D. L., Sascău, R. A., Balasanian, M., Macovei, L., Arsenescu-Georgescu, C., & Stătescu, C. (2022). Beta-Blocker-Related Atrioventricular Conduction Disorders—A single tertiary referral Center experience. Medicina-lithuania, 58(2), 320. https://doi.org/10.3390/medicina58020320
  42. Westergaard, L. M., Alhakak, A., Rørth, R., Fosbøl, E. L., Kristensen, S. L., Svendsen, J. H., Graff, C., Jb, N., Gislason, G., Køber, L., Torp‐Pedersen, C., Lee, C. J., & Weeke, P. (2023). Ventricular rate in atrial fibrillation and the risk of heart failure and death. Europace, 25(5). https://doi.org/10.1093/europace/euad088
  43. Grubb A, Mentz RJ. Pharmacological management of atrial fibrillation in patients with heart failure with reduced ejection fraction: review of current knowledge and future directions. Expert Rev Cardiovasc Ther. 2020 Feb;18(2):85-101
  44. David, M. N. V. (2023, January 19). Digoxin. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK556025/
  45. Alqahtani, M. S., Kazi, M., Alsenaidy, M. A., & Ahmad, M. Z. (2021). Advances in oral drug delivery. Frontiers in Pharmacology, 12. https://doi.org/10.3389/fphar.2021.618411
  46. Cheng, L., & Wong, H. (2020). Food effects on oral drug absorption: Application of Physiologically Based Pharmacokinetic Modeling as a predictive tool. Pharmaceutics, 12(7), 672. https://doi.org/10.3390/pharmaceutics12070672
  47. Herman, T. F. (2023, November 3). First-Pass effect. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK551679/
  48. Mar, P. L., Horbal, P., Chung, M. K., Dukes, J. W., Ezekowitz, M. D., Lakkireddy, D., Lip, G. Y. H., Miletello, M., Noseworthy, P. A., Reiffel, J. A., Tisdale, J. E., Olshansky, B., & Gopinathannair, R. (2022). Drug interactions affecting antiarrhythmic drug use. Circulation: Arrhythmia and Electrophysiology, 15(5). https://doi.org/10.1161/circep.121.007955
  49. Florek, J. B. (2023, November 12). Amiodarone. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK482154/
  50. Dean, L. (2017, April 4). Propafenone therapy and CYP2D6 genotype. Medical Genetics Summaries – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK425391/
  51. Rollason, V., Lloret‐Linares, C., Lorenzini, K. I., Daali, Y., Gex‐Fabry, M., Piguet, V., Besson, M., Samer, C. F., & Desmeules, J. A. (2020). Evaluation of phenotypic and genotypic variations of drug metabolizing enzymes and transporters in chronic pain patients facing adverse drug reactions or Non-Response to analgesics: a retrospective study. Journal of Personalized Medicine, 10(4), 198. https://doi.org/10.3390/jpm10040198
  52. Chu, R. (2020, October 10). Intravenous lidocaine infusion for the management of early postoperative pain: A Comprehensive review of controlled trials. PubMed Central (PMC). https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7901134/
  53. Weibel, S., Jelting, Y., Pace, N. L., Helf, A., Eberhart, L., Hahnenkamp, K., Hollmann, M. W., Poepping, D., Schnabel, A., & Kranke, P. (2018). Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery in adults. The Cochrane Library, 2018(6). https://doi.org/10.1002/14651858.cd009642.pub3
  54. Larson, J., Rich, L., Deshmukh, A., Judge, E. C., & Liang, J. J. (2022). Pharmacologic Management for Ventricular arrhythmias: Overview of Anti-Arrhythmic Drugs. Journal of Clinical Medicine, 11(11), 3233. https://doi.org/10.3390/jcm11113233
  55. Tun, M. M., Pandey, S., Adhikari, S., Mainali, A., Thapa, A., Bisural, R., Bista, P. B., Htet, S. Y., Chhetri, B., & Panigrahi, K. (2023). Amiodarone-Induced liver attenuation on CT scan: alarming signal for toxicity and prompt discontinuation. Cureus. https://doi.org/10.7759/cureus.39844
  56. Sasi, N., MD. (2024). Medscape Registration. https://emedicine.medscape.com/article/813046-overview?form=fpf
  57. Ohyama, K., Nakajima, M., Suzuki, M., Shimada, N., Yamazaki, H., & Yokoi, T. (2000). Inhibitory effects of amiodarone and its N‐deethylated metabolite on human cytochrome P450 activities: Prediction of in vivo drug interactions. British Journal of Clinical Pharmacology, 49(3), 244–253. https://doi.org/10.1046/j.1365-2125.2000.00134.x
  58. Beavers, C. J., Rodgers, J. E., Bagnola, A., Beckie, T. M., Campia, U., Di Palo, K. E., Okwuosa, T. M., Przespolewski, E., & Dent, S. (2022). Cardio-Oncology Drug Interactions: A scientific statement from the American Heart Association. Circulation, 145(15). https://doi.org/10.1161/cir.0000000000001056
  59. Kamaraju, S., Mohan, M., Zaharova, S., Wallace, B., McGraw, J., Lokken, J., Tierney, J. F., Weil, E., Fatunde, O., & Brown, S. (2021). Interactions between cardiology and oncology drugs in precision cardio-oncology. Clinical Science, 135(11), 1333–1351. https://doi.org/10.1042/cs20200309
  60. Chakraborty, R. K. (2023, July 28). Calcium channel blocker toxicity. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK537147/
  61. Konieczny, K. M., & Dorian, P. (2019). Clinically important Drug–Drug interactions between antiarrhythmic drugs and anticoagulants. The Journal of Innovations in Cardiac Rhythm Management, 0(3), 3552–3559. https://doi.org/10.19102/icrm.2019.100304
  62. Ferri, N., Colombo, E., Tenconi, M., Baldessin, L., & Corsini, A. (2022). Drug-Drug interactions of direct oral anticoagulants (DOACs): From pharmacological to clinical practice. Pharmaceutics, 14(6), 1120. https://doi.org/10.3390/pharmaceutics14061120
  63. Hügl, B., Horlitz, M., Fischer, K., & Kreutz, R. (2023). Clinical significance of the rivaroxaban–dronedarone interaction: insights from physiologically based pharmacokinetic modelling. European Heart Journal Open, 3(1). https://doi.org/10.1093/ehjopen/oead004
  64. Lucà, F., Oliva, F., Abrignani, M. G., Di Fusco, S. A., Parrini, I., Canale, M. L., Giubilato, S., Cornara, S., Nesti, M., Rao, C. M., Pozzi, A., Binaghi, G., Maloberti, A., Ceravolo, R., Bisceglia, I., Rossini, R., Temporelli, P. L., Amico, A., Calvanese, R., . . . Gulizia, M. M. (2023). Management of Patients Treated with Direct Oral Anticoagulants in Clinical Practice and Challenging Scenarios. Journal of Clinical Medicine, 12(18), 5955. https://doi.org/10.3390/jcm12185955
  65. Wiggins, B. S., Dixon, D. L., Neyens, R., Page, R. L., & Gluckman, T. J. (2020). Select Drug-Drug interactions with direct oral anticoagulants. Journal of the American College of Cardiology, 75(11), 1341–1350. https://doi.org/10.1016/j.jacc.2019.12.068
  66. Tovia-Brodie, O. (2020). Pharmacological therapy in Brugada syndrome. Radcliffe Cardiology. https://www.aerjournal.com/articles/pharmacological-therapy-brugada-syndrome
  67. Sanchez‐Nadales, A., Anampa-Guzmán, A., & Khan, A. (2019). Disopyramide for hypertrophic cardiomyopathy. Cureus. https://doi.org/10.7759/cureus.4526
  68. Blotner, M., Betageri, O., Miles, W. M., & Kong, X. (2022). WorkUp for Suspected Brugada Syndrome: Two case reports for the general practitioner. Cureus. https://doi.org/10.7759/cureus.21921
  69. Chhabra, L. (2023, August 7). Wolff-Parkinson-White Syndrome. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK554437/
  70. Bohnen, M. S., Peng, G., Robey, S., Terrenoire, C., Iyer, V., Sampson, K. J., & Kass, R. S. (2017). Molecular Pathophysiology of Congenital Long QT Syndrome. Physiological Reviews, 97(1), 89–134. https://doi.org/10.1152/physrev.00008.2016
  71. Narayanan, K. (2020). Strategies for rhythm control in Atrial fibrillation. Indian Journal of Clinical Cardiology, 1(2), 94–107. https://doi.org/10.1177/2632463620936018
  72. Arunachalam, K. (2023, August 8). Flecainide. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK542291/
  73. Sreenivas, S. (2021, March 3). Beta-Blocker Medications for AFIB. WebMD. https://www.webmd.com/heart-disease/atrial-fibrillation/beta-blocker-medications-afib
  74. Viskin, S., Chorin, E., Viskin, D., Hochstadt, A., Schwartz, A. L., & Rosso, R. (2021). Polymorphic Ventricular Tachycardia: Terminology, mechanism, diagnosis, and emergency therapy. Circulation, 144(10), 823–839. https://doi.org/10.1161/circulationaha.121.055783
  75. Muser, D., Tritto, M., Mariani, M. V., Di Monaco, A., Compagnucci, P., Accogli, M., De Ponti, R., & Guarracini, F. (2021). Diagnosis and treatment of idiopathic Premature ventricular contractions: A stepwise approach based on the site of origin. Diagnostics, 11(10), 1840. https://doi.org/10.3390/diagnostics11101840
  76. Khachatryan, A., Merino, J. L., De Abajo, F. J., Botto, G. L., Kirchhof, P., Breithardt, G., Stambler, B. S., Abenhaim, L., & Grimaldi-Bensouda, L. (2021). International cohort study on the effectiveness of dronedarone and other antiarrhythmic drugs for atrial fibrillation in real-world practice (EFFECT-AF). Europace, 24(6), 899–909. https://doi.org/10.1093/europace/euab262
  77. Clinical use of dofetilide – UpToDate. (2024). UpToDate. https://www.uptodate.com/contents/clinical-use-of-dofetilide
  78. Mubarik, A. (2022, June 10). Sotalol. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK534832/
  79. Szymanski, M. W. (2023, April 3). Ibutilide. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK526021/
  80. Phillips, C. T., Wang, J., Celi, L. A., Zhang, Z., & Feng, M. (2019). Association of hypokalemia with an increased risk for medically treated arrhythmias. PLOS ONE, 14(6), e0217432. https://doi.org/10.1371/journal.pone.0217432
  81. PROPAFENONE HYDROCHLORIDE TABLETS. (2024). https://dailymed.nlm.nih.gov/dailymed/fda/fdaDrugXsl.cfm?setid=a7c8f090-c48c-44f7-9973-2cf4b491e35c&type=display
  82. Intro_to_antiarrhythmics [TUSOM | Pharmwiki]. (2024). https://tmedweb.tulane.edu/pharmwiki/doku.php/intro_to_antiarrhythmics
  83. Singh, S. (2023, August 28). Adenosine. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK519049/
  84. Alobaida, M., & Alrumayh, A. (2021). Rate control strategies for atrial fibrillation. Annals of Medicine, 53(1), 682–692. https://doi.org/10.1080/07853890.2021.1930137
  85. Fatima, N., Mandava, K., Khatoon, F., Badar, J., Begum, S. F., Narasimhan, C., Daljeet, K., & Mohammed, W. (2022). CLInical Profile and Side Effects of chronic use of oral Amiodarone in cardiology outpatient department (CLIPSE-A Study)- A prospective observational study. Annals of Medicine and Surgery, 80. https://doi.org/10.1016/j.amsu.2022.104167
  86. Dunn, J. (2023, February 13). Physiology, adenosine triphosphate. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK553175/
  87. Lavonas, E. J., Akpunonu, P., Arens, A. M., Babu, K. M., Cao, D., Hoffman, R. S., Hoyte, C., Mazer‐Amirshahi, M., Stolbach, A., St‐Onge, M., Thompson, T. M., Wang, G. S., Hoover, A. V., & Drennan, I. R. (2023). 2023 American Heart Association Focused Update on the Management of Patients with Cardiac Arrest or Life-Threatening Toxicity Due to Poisoning: An Update to the American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation, 148(16). https://doi.org/10.1161/cir.0000000000001161
  88. Rehman, R. (2023, May 1). Digitalis toxicity. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK459165/
  89. Pincus M. (2016). Management of digoxin toxicity. Australian prescriber, 39(1), 18–20. https://doi.org/10.18773/austprescr.2016.006
  90. Crosby, J., Bhopalwala, H., Kharawala, A., Dewaswala, N., Ganti, S. S., & Bhopalwala, A. (2021). Refractory Torsades de Pointes Due to Dofetilide Overdose. Journal of Investigative Medicine High Impact Case Reports, 9, 232470962110564. https://doi.org/10.1177/23247096211056492
  91. Khalid, M. M. (2023, July 28). Beta-Blocker toxicity. StatPearls – NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK448097/
  92. Bologa, C., Lionte, C., Popescu, A., Sorodoc, V., & Sorodoc, L. (2021). First Case of Acute Poisoning with Amiodarone and Flecainide in Attempted Suicide Successfully Managed with Lipid Emulsion Therapy in the Emergency Department: Case Report and Literature Review. Healthcare (Basel, Switzerland), 9(6), 671. https://doi.org/10.3390/healthcare9060671
  93. Lei, M., Wu, L., Terrar, D. A., & Huang, C. L. (2018). Modernized Classification of Cardiac Antiarrhythmic Drugs. Circulation, 138(17), 1879–1896. https://doi.org/10.1161/CIRCULATIONAHA.118.035455
  94. Hakkola, J., Hukkanen, J., Turpeinen, M., & Pelkonen, O. (2020). Inhibition and induction of CYP enzymes in humans: an update. Archives of Toxicology, 94(11), 3671–3722. https://doi.org/10.1007/s00204-020-02936-7

 

Disclaimer:

Use of Course Content. The courses provided by NCC are based on industry knowledge and input from professional nurses, experts, practitioners, and other individuals and institutions. The information presented in this course is intended solely for the use of healthcare professionals taking this course, for credit, from NCC. The information is designed to assist healthcare professionals, including nurses, in addressing issues associated with healthcare. The information provided in this course is general in nature and is not designed to address any specific situation. This publication in no way absolves facilities of their responsibility for the appropriate orientation of healthcare professionals. Hospitals or other organizations using this publication as a part of their own orientation processes should review the contents of this publication to ensure accuracy and compliance before using this publication. Knowledge, procedures or insight gained from the Student in the course of taking classes provided by NCC may be used at the Student’s discretion during their course of work or otherwise in a professional capacity. The Student understands and agrees that NCC shall not be held liable for any acts, errors, advice or omissions provided by the Student based on knowledge or advice acquired by NCC. The Student is solely responsible for his/her own actions, even if information and/or education was acquired from a NCC course pertaining to that action or actions. By clicking “complete” you are agreeing to these terms of use.

 

Complete Survey

Give us your thoughts and feedback

Want to earn credit for this course? Sign up (new users) or Log in (existing users) to complete this course for credit and receive your certificate instantly.