Antiarrhythmic Electrophysiology and Pharmacotherapy

CHAPTER 40 Antiarrhythmic Electrophysiology and Pharmacotherapy




CARDIAC ARRHYTHMIAS continue to be a significant cause of morbidity and mortality in the developed world and add to the complexity of management of critically ill patients. Over the past 2 decades treatment strategies have expanded to include other approaches such as catheter ablation and implantable cardiac defibrillators. Although these strategies have demonstrated significant superiority over antiarrhythmics, they have not eliminated the problem of cardiac arrhythmias. Furthermore, these approaches can be difficult to apply to patients in the acute setting. Antiarrhythmic agents continue to be a vital adjunct to these therapeutic approaches.


Despite the important role that antiarrhythmics play to combat cardiac arrhythmias, it has been difficult to overcome the stigma associated with these drugs. This stigma has stemmed from earlier published trials such as the CAST and CAST II trials evaluating class IC antiarrhythmics and the SWORD trial evaluating sotalol in patients with coronary artery disease and left ventricular dysfunction. These trials demonstrated increased mortality, significant side effects, and marginal efficacy.1,2,3 The Achilles heel of these agents is their propensity for proarrhythmia that results in more dangerous arrhythmias than the ones they were initially intended to treat. This led to a re-evaluation of the wisdom of using relatively simple agents that took aim at single molecular targets, such as sodium or potassium channels. Subsequently, researchers pursued the development of newer antiarrhythmic agents that target multiple molecules or complex arrhythmogenic pathways, such as amiodarone or d,l-sotalol. There was a renewed interest and emphasis on the use of β-blockers, which do not specifically target ion channels. In addition there has been significant interest in understanding the genetic basis of susceptibility to serious arrhythmias in individuals with genetic syndromes such as long QT and Brugada syndromes.


Despite their potentially serious side effects, antiarrhythmics continue to be a valuable tool when used appropriately and in a targeted manner. The aim of this chapter is to provide a clear and concise overview of the physiologic basis and applicable pharmacology that makes these drugs useful, and to discuss the ones most clinically relevant to the management of the acutely ill patient.




His-Purkinje Action Potential


The His-Purkinje action potential can be conceptually divided into periods of depolarization, repolarization, and resting states. However, the action potential is traditionally divided into five different phases (phases 0-4) to describe the activity of various ion channels that bring about any of these three states (Fig. 40-1, A and B). Phase 4 corresponds to the resting state when the cell is not being stimulated and is ready for subsequent depolarization. Phase 0 corresponds to depolarization of the myocardial cell. It initiates a cascade of events involving the influx and efflux of multiple ions, leading to phases 1-3, manifesting in repolarization and refractoriness.









SA Node and AV Node Action Potential


SA and AV nodal action potentials are very similar with only minor differences between them in phase 0. They are significantly different from fast response tissue’s action potentials previously described. These slow response tissue action potentials are divided into three phases instead of five (Fig. 40-2, A and B). Phase 4 is the spontaneous depolarization (pacemaker potential) phase that triggers the action potential once the membrane potential reaches threshold (between −40 and −30 mV). Phase 0 is the depolarization phase of the action potential. This is followed by phase 3 repolarization. Once the cell is completely repolarized at about −60 mV, the cycle is spontaneously repeated.








Classification



Vaughan-Williams


This is currently the most widely used classification of antiarrhythmic drugs (Table 40-1). This classification was initially introduced by Vaughan Williams6,7 and was based on the electrophysiologic effects of antiarrhythmics. It was later modified by Harrison8 with the recognition that drugs in the same class have different potencies and that the same drug may exert multiple class effects.


Table 40–1 Vaughan-Williams Classification of Antiarrhythmics and Their Mechanism of Action







































Class Mechanism Drugs
I Na+ channel blockers  
A Slows conduction velocity (Vmax) and prolongs action potential. Results in decreased conductivity and increased refractoriness. Quinidine, procainamide, disopyramide
B Minimal effect on conduction velocity (Vmax) and shortens action potential duration. Results in decreased refractoriness. Lidocaine, mexiletine
C Significant slowing of conduction velocity (Vmax), minimal effect on action potential duration. Results in decreased conductivity and no change in refractoriness. Flecainide, propafenone, moricizine
II

Atenolol, esmolol, propranolol
III

Amiodarone, sotalol, dofetilide, ibutilide, azimilide
IV

Verapamil, diltiazem (non-dihydropyridine)
V

Digoxin, adenosine

Amiodarone has effects across all classes, blocking Na+ channels in depolarized tissues, but is also capable of affecting Ca2+ and K+ channels and adrenergic receptors. This makes it an extremely versatile drug capable of affecting a wide variety of arrhythmias. Sotalol has a significant β-blocker effect in addition to its class III action. Ibutilide can also enhance the slow delayed sodium current. Although moricizine has diverse effects across different classes, this drug is not currently used because it has been demonstrated to increase mortality.



Class I Antiarrhythmics and Use Dependence


Class I drugs are all Na+ channel blockers with varying potency and are classified based on their effect on the action potential upstroke or Vmax and therefore their ability to alter conduction velocity. They can prolong it, shorten it, or have no net effect (Fig. 40-3). Their different potency is due to variable rates of binding and dissociation from the channel receptor.9 Class IC are the most potent Na+ channel blockers because they have the slowest binding and dissociation from the receptor; class IA are the least potent (fastest binding and dissociation); and class IB are moderately potent. Faster heart rates allow less time for drug-receptor dissociation, resulting in an increased total number of blocked receptors and more effective antiarrhythmic action. This is known as “use dependence” and is most noted with class IC agents.10 These effects can result in prolonged conduction velocity and manifest by widening of the QRS complex on the surface electrocardiogram (ECG), especially during tachycardia.




Class III Antiarrhythmics and Reverse Use Dependence


Class III antiarrhythmic agents extend the plateau phase of the action potential by blocking K+ channels. They effectively prolong repolarization and the action potential duration with a resultant increase in refractoriness without a change in conductivity (Fig. 40-4). This manifests as prolongation of the Q–T interval on the surface ECG. These effects are most pronounced during slow heart rates. This is known as “reverse use dependence” and thus, longer Q–T intervals are noted at slower heart rates.11 This provides the potential for dangerous arrhythmias such as torsades de pointes.12 This is known as proarrhythmia and is one of the most serious, life-threatening side effects associated with antiarrhythmic drug use. Amiodarone, however, appears to be the exception, with proarrhythmia being reported only uncommonly.13




Class II Antiarrhythmics


These drugs act by competitive inhibition of the β-adrenergic receptor, and largely affect the SA and AV node. Yet they also have mild Na+ channel inhibitory effects. There is a preponderance of data demonstrating the antiarrhythmic properties of β-blockers. β-blockers have played an increasingly important role in increasing survival in patients with coronary heart disease and congestive heart failure, along with adjunctive therapy to implantable cardiac defibrillators (ICDs) by decreasing the incidence of sudden cardiac death (SCD). Although not all of the beneficial effects of β-blockers are understood it is likely that they essentially work by reversing or preventing the proarrhythmic actions of sympathetic activity.14 These include increased automaticity because of enhanced phase 4 depolarization in the SA and AV nodes, increased membrane excitability in phase 2 and 3 of the His-Purkinje action potential, increased Vmax, and increased delayed afterpotentials, which can lead to increased triggered activity type arrhythmias. They are therefore most effective in tissue under intense adrenergic stimulation (e.g., in ischemia) and it is not surprising that their effects are most obvious in patients having acute myocardial infarction and decompensated heart failure.





Sicilian Gambit


Although the merits of the Vaughan-Williams classification are its simplicity and wide recognition, providing a useful mode for communication regarding the use of antiarrhythmics, it does not take into account several complexities. These include drugs’ ability to exert effects that cross into different classes, having variable potencies within the same class, and causing other effects such as changes in metabolism, autonomic stimulation, or hemodynamics. The Sicilian gambit17 (Table 40-2) was introduced as a method of rationalizing the approach to the use of antiarrhythmics. Although not widely known and far more complex in its original format, it is included here for its emphasis on approaching antiarrhythmic drug choice based on the mechanism of each individual arrhythmia and the perceived “vulnerable parameter” that can be affected to terminate it.




Pharmacology


Pharmacologic concerns of drug therapy can be divided into two main disciplines. The first is pharmacokinetics, which describes the process of drug delivery to its target site. It encompasses the processes of drug absorption, distribution, metabolism, and elimination. This can be summarized in the relationship between drug dose and plasma concentration over time. This should not be confused with pharmacodynamics, which describes target-specific drug interaction, and the resultant downstream whole body effects. This can be thought of as the relationship between drug concentration and magnitude of drug effect.


Obviously, both of these processes can then be impacted by various factors such as drug absorption, bioavailability, volume of distribution, drug clearance, elimination, and half-life. Due to the complexity of drug transport and activity, genetic variability, and the heterogeneous and often abnormal metabolic milieu of the critically ill patient, significant interpatient variability can be expected in their response to the same drugs.




Distribution


A drug is distributed to the best perfused tissues first, termed the central compartment, such as the heart, lungs, and brain. Subsequently, it reaches less perfused tissues such as the skin and muscle—the peripheral compartment. Some drugs such as amiodarone have very slow distribution to tissues such as adipose, termed the deep compartment, which prevent it from reaching a steady state until these tissues are saturated.


The volume of distribution (Vd) of a particular drug refers to a theoretical space or volume into which the drug is distributed and can be used to describe the relationship between drug dose and plasma concentration. This can in turn be affected by changes in the actual plasma volume reflected by tissue perfusion, as can be noted in cases of congestive heart failure or shock. Alternatively, tissue affinity, fraction of drug bound to proteins (only the free portion of a drug produces the desired effect) and degree of lipid or water solubility can all play a role in altering the volume of distribution.


A classic example is lidocaine administration in heart failure patients. The Vd may be reduced by 40%18 because of decreased perfusion and therefore unless lidocaine bolus doses are reduced in these patients, lidocaine toxicity can result.


Most antiarrhythmics are bound to α1-glycoprotein. This protein’s concentration increases with acute and critical illness (such as shock, trauma, or severe bacterial infection). The result may be decreased effectiveness of the antiarrhythmic agent despite an unchanged plasma concentration, as the fraction of unbound (free) active drug is smaller.



Metabolism and Elimination


Most antiarrhythmics are metabolized hepatically by the cytochrome P450 system, and many undergo extensive first pass metabolism. This accounts for the large ratio of oral to intravenous doses of such medicines as verapamil, metoprolol, or propafenone. Others such as lidocaine are so completely eliminated by the first pass effect that oral dosage is useless.19


Metabolism of the parent drug produces metabolites that may or may not be active. Furthermore, these metabolites may have similar or different effects than the parent drug. Classic examples include amiodarone metabolism to desethylamiodarone, which accounts for a persistent drug effect beyond the drug’s half-life. Procainamide, however, a class Ia agent, is metabolized to N-acetylprocainamide (NAPA), which has a predominant class III effect.


A drug’s half-life describes the time required to reduce a plasma drug concentration by 50%. This may be secondary to either drug metabolism and elimination or redistribution. It typically requires five half-lives to eliminate a drug from the plasma completely and five half lives to achieve a steady state. Since only three doses are required to achieve greater than 90% steady state plasma concentrations, drug loading doses should only be used when such an interval is clinically unacceptable such as in life-threatening arrhythmias in critically ill patients. In such situations, a loading dose may be given to achieve desirable plasma concentration levels much faster.20



Antiarrhythmics of Clinical Relevance in the CCU


Although detailed accounts of every antiarrhythmic are available elsewhere, the focus of this review is to discuss the antiarrhythmics that have the most clinical relevance to the management of the critically ill patient in the acute care setting. Older medications no longer in current use and those that cannot be administered intravenously or are contraindicated in critical illness are not discussed (Table 40-3).



Jun 4, 2016 | Posted by in CRITICAL CARE | Comments Off on Antiarrhythmic Electrophysiology and Pharmacotherapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access