Atrial Fibrillation




Abstract


Atrial fibrillation (AF) is the most common sustained arrhythmia encountered in clinical practice. AF is a progressive disease and is independently associated with significantly increased morbidity and mortality.


The pathogenesis of AF remains incompletely understood and is believed to be complex, multifactorial, and variable in different individuals. The factors responsible for the onset of AF include triggers that induce the arrhythmia and a receptive substrate that sustains it. Triggering foci of rapidly firing cells within the sleeves of atrial myocytes extending into the pulmonary veins have been shown to be the underlying mechanism in most cases of paroxysmal AF.


Management of AF should be aimed at identifying and treating underlying causes of the arrhythmia, as well as reducing symptoms, improving quality of life, and preventing cardiovascular morbid­ity and mortality associated with AF. There are four main issues that must be addressed in the treatment of AF: (1) prevention of systemic embolization; (2) ventricular rate control; (3) restoration and maintenance of NSR; and (4) risk factor modification.


Catheter ablation of AF provides higher efficacy with comparable safety as antiarrhythmic drug therapy and is recommended in patients with symptomatic AF as a second-line treatment after failure of or intolerance to class I or III antiarrhythmic drug therapy, and as a first-line treatment in selected AF patients. Electrical isolation of all pulmonary veins is recommended during all AF ablation procedures. Ablation of extrapulmonary vein focal triggers is recommended if a reproducible trigger that initiates AF is identified at the time of an AF ablation procedure. Left atrial posterior wall isolation may be considered for initial or repeat ablation of persistent or longstanding persistent AF. The usefulness of other substrate-based mapping and ablation approaches (including linear ablation of various sites, ablation of ganglionated plexuses, complex fractionated atrial electrogram ablation, focal impulse and rotor modulation, and voltage-guided ablation) as an initial or repeat ablation strategy for persistent or longstanding persistent AF is not well established.




Keywords

atrial fibrillation, pulmonary vein isolation, ganglionated plexuses, complex fractionated atrial electrograms, focal impulse and rotor modulation, anticoagulation, left atrial appendage closure

 






  • Outline



  • Pathophysiology, 422




    • Classification of Atrial Fibrillation, 422



    • Mechanism of Atrial Fibrillation, 423



    • Substrate for Atrial Fibrillation, 427



    • Atrial Remodeling in Atrial Fibrillation, 429



    • Role of Autonomic Nervous System in Atrial Fibrillation, 430



    • Role of the Pulmonary Veins in Atrial Fibrillation, 430



    • Genetics in Atrial Fibrillation, 433




  • Epidemiology, 433




    • Atrial Fibrillation Risk Prediction, 434



    • Clinical Risk Factors Predisposing to Atrial Fibrillation, 434



    • Drug-Induced Atrial Fibrillation, 438



    • Postoperative Atrial Fibrillation, 439




  • Clinical Presentation, 439




    • Symptomatic Atrial Fibrillation, 439



    • Atrial Fibrillation Symptom Scales, 440



    • Silent Atrial Fibrillation, 440



    • Device-Detected Atrial Fibrillation, 440




  • Risk of Thromboembolism, 441




    • Stroke Risk Stratification, 442



    • Bleeding Risk Stratification, 443




  • Initial Evaluation, 444




    • Diagnostic Cardiac Testing, 444



    • Laboratory Testing, 445



    • Electrophysiological Testing, 445



    • Other Diagnostic Tests, 445



    • Screening for Atrial Fibrillation, 445




  • Principles of Management, 445




    • Prevention of Systemic Embolization, 445



    • Rate Control, 448



    • Rhythm Control, 449



    • Upstream Therapy, 455



    • Risk Factor Management, 456



    • Management of Postoperative Atrial Fibrillation, 457




  • Electrocardiographic Features, 457




    • Atrial Activity, 457



    • Atrioventricular Conduction During Atrial Fibrillation, 458



    • QRS Morphology, 461




  • Catheter Ablation of Atrial Fibrillation, 462




    • Evolution of Catheter Ablation Approaches for Atrial Fibrillation, 462



    • Periprocedural Management, 463



    • Technical Aspects Common to Different Methods of Ablation, 465




  • Focal Ablation of Pulmonary Vein Triggers, 467




    • Rationale, 467



    • Identification of Arrhythmogenic Pulmonary Veins, 467



    • Mapping Pulmonary Vein Ectopy, 467



    • Target of Ablation, 470



    • Ablation Technique, 470



    • Endpoints of Ablation, 470



    • Outcome, 470




  • Segmental Ostial Pulmonary Vein Isolation, 471




    • Rationale, 471



    • Circumferential Mapping of Pulmonary Vein Potentials, 471



    • Target of Ablation, 480



    • Ablation Technique, 480



    • Endpoints of Ablation, 480



    • Outcome, 483




  • Circumferential Antral Pulmonary Vein Isolation, 483




    • Rationale, 483



    • Identification of Pulmonary Vein Antra, 483



    • Target of Ablation, 485



    • Ablation Technique, 487



    • Endpoints of Ablation, 493



    • Outcome, 493




  • Circumferential Left Atrial Ablation, 494




    • Rationale, 494



    • Electroanatomic Mapping, 494



    • Target of Ablation, 495



    • Ablation Technique, 495



    • Endpoints of Ablation, 496



    • Outcome, 497




  • Linear Atrial Ablation, 497




    • Left Atrial Roof Line, 497



    • Left Atrial Posterior Wall Isolation, 498



    • Lateral Mitral Isthmus Line, 498



    • Cavotricuspid Isthmus Line, 500




  • Focal Impulse and Rotor Mapping, 501




    • Rationale, 501



    • Focal Impulse and Rotor Modulation Mapping, 501



    • Target of Ablation, 501



    • Ablation Technique, 501



    • Endpoints of Ablation, 502



    • Outcome, 503




  • Ablation of Complex Fractionated Atrial Electrograms, 504




    • Rationale, 504



    • Mapping Complex Fractionated Atrial Electrograms, 504



    • Target of Ablation, 506



    • Ablation Technique, 506



    • Endpoints of Ablation, 506



    • Outcome, 507




  • Pulmonary Vein Denervation, 507




    • Rationale, 507



    • Localization of Ganglionated Plexuses, 507



    • Target of Ablation, 508



    • Ablation Technique, 508



    • Endpoints of Ablation, 509



    • Outcome, 509




  • Ablation of Non–Pulmonary Vein Triggers, 509




    • Rationale, 509



    • Mapping of Non–Pulmonary Vein Triggers, 509



    • Mapping and Ablation of the Ligament of Marshall, 510



    • Electrical Isolation of the Superior Vena Cava, 512



    • Electrical Isolation of the Coronary Sinus, 513



    • Electrical Isolation of the Left Atrial Appendage, 515



    • Outcome, 515




  • Voltage-Guided Substrate Modification, 515




    • Rationale, 515



    • Target of Ablation, 515



    • Ablation Technique, 515



    • Endpoint of Ablation, 516



    • Outcome, 516




  • Outcome and Efficacy of Catheter Ablation of Atrial Fibrillation, 516




    • Success Rates, 517



    • Recurrence of Atrial Tachyarrhythmias, 519



    • Atrial Tachycardia and Flutter Following Ablation of Atrial Fibrillation, 520



    • Complications of Catheter Ablation of Atrial Fibrillation, 522




  • Recommendations and Controversies, 525




    • Determination of the Necessity of Pulmonary Vein Electrical Isolation, 525



    • Determination of the Necessity of Adjunctive Substrate Modification, 525




  • Atrioventricular Junction Ablation, 526




    • Rationale, 526



    • Target of Ablation, 527



    • Ablation Technique, 527



    • Endpoints of Ablation, 528



    • Outcome, 528




  • Atrioventricular Nodal Modification, 530




    • Rationale, 530



    • Target of Ablation, 530



    • Ablation Technique, 531



    • Endpoints of Ablation, 531



    • Outcome, 531




  • Percutaneous Left Atrial Appendage Device Closure, 531




    • Left Atrial Appendage Anatomy, 532



    • Left Atrial Appendage Imaging, 532



    • Left Atrial Appendage Catheterization, 534



    • Watchman Device, 534



    • Amplatzer Cardiac Plug, 535




  • Percutaneous Left Atrial Appendage Ligation, 537




    • Left Atrial Appendage Imaging, 537



    • Device Specification, 537



    • Epicardial Access, 537



    • Endocardial Access, 537



    • Connecting the Epicardial and Endocardial Magnet-Tipped Guidewires, 538



    • Snaring the Left Atrial Appendage, 538



    • Postoperative Management, 538



    • Outcome, 538




  • Surgical Left Atrial Appendage Exclusion, 540




Pathophysiology


Classification of Atrial Fibrillation


Atrial fibrillation (AF) has been described as lone, idiopathic, nonvalvular, valvular, paroxysmal, persistent, or permanent. Each of these classifications has implications regarding mechanisms as well as response to therapy. At the initial detection of AF, it is impossible to know the subsequent pattern of duration and frequency of recurrences. Thus a designation of first-detected episode of AF is made on the initial diagnosis, irrespective of the duration of the arrhythmia. When the patient has experienced two or more episodes, AF is classified as recurrent.


After termination of an episode of AF, the rhythm can be classified as paroxysmal or persistent ( Table 15.1 ). Paroxysmal AF is characterized by self-terminating episodes that generally last less than 7 days. Persistent AF lasts longer than 7 days and often requires electrical or pharmacological cardioversion. Subcategories of persistent AF (according to arrhythmia duration) include early persistent AF (defined as AF that is sustained beyond 7 days but is less than 3 months in duration) and longstanding persistent AF (defined as AF that is sustained longer than 1 year but is being considered for ablation). Permanent AF refers to AF in which cardioversion has failed or AF that has been sustained for more than 1 year and further attempts to restore normal sinus rhythm (NSR) were unsuccessful or have been abandoned.



TABLE 15.1

Classifications of Atrial Fibrillation






















AF Pattern Definition
First diagnosed AF AF that has not been diagnosed before, irrespective of the duration of the arrhythmia or the presence and severity of AF-related symptoms.
Paroxysmal AF Self-terminating, in most cases within 48 hours. Some AF paroxysms may continue for up to 7 days. a
Persistent AF AF that lasts longer than 7 days, including episodes that are terminated by cardioversion, either with drugs or by direct current cardioversion, after 7 days or more.
Longstanding persistent AF Continuous AF lasting for ≥1 year when it is decided to adopt a rhythm control strategy.
Permanent AF AF that is accepted by the patient (and physician). Hence, rhythm control interventions are, by definition, not pursued in patients with permanent AF. Should a rhythm control strategy be adopted, the arrhythmia would be re-classified as “longstanding persistent AF.”

AF, Atrial fibrillation.

From Kirchhof P, Benussi S, Kotecha D, et al. 2016 ESC guidelines for the management of atrial fibrillation developed in collaboration with EACTS. Eur Heart J . 2016;37:2893–2962.

a The distinction between paroxysmal and persistent AF is often not made correctly without access to long-term monitoring. Hence, this classification alone is often insufficient to select specific therapies. If both persistent and paroxysmal episode are present, the predominant pattern should guide the classification.



Although useful, this arbitrary classification does not account for all presentations of AF and overlap occurs. Paroxysmal AF often progresses to longer, non-self-terminating episodes. In addition, the pattern of AF can change in response to treatment. AF that has been persistent can become paroxysmal with antiarrhythmic drug therapy, and AF that had been permanent can potentially be cured or made paroxysmal by surgical or catheter-based ablation. Furthermore, the distinction between persistent and permanent AF is not only a function of the underlying arrhythmia but also a reflection of the clinical pragmatism of the patient and physician. The severity of symptoms associated with AF, anticoagulation status, and patient preference all affect the decision of whether and when cardioversion will be attempted. This decision would then affect the duration of sustained AF and could lead to a diagnosis of persistent or permanent AF. Furthermore, significant discrepancies exist between the clinical AF classification and the objective cardiac device-derived assessments of AF temporal persistence (in AF patients with pacemakers or defibrillators) ( eFig. 15.1 ). Patients within the same clinical class (“paroxysmal” or “persistent” AF) are highly heterogeneous with regard to AF temporal persistence, arrhythmia burden, and stages of disease.





eFig. 15.1


Clinical Versus Cardiac Device-Derived Classification of Atrial Fibrillation (AF) .

(A) Distribution of the AF burden in the clinical classification groups (paroxysmal, persistent). (B) The probability of being in either of the clinical classification groups at any given AF burden level (conditional density plot). For example, at an AF burden of 0.2, the probability of being classified as “paroxysmal” or “persistent” was approximately 0.8 and 0.2, respectively. As the AF burden increases, intuitively one would expect that the probability of being classified as “paroxysmal” AF decreases and the probability of being classified as “persistent” increases. Unexpectedly, at AF burdens of 0.9, the probability of being classified as “paroxysmal” or “persistent” was also approximately 0.8 and 0.2, respectively, showing the disconnection between clinical classification and temporal persistence. Even at very high burdens the vast majority of patients were classified as having “paroxysmal” AF. (C) Distribution of the atrial fibrillation (AF) burden in the device-derived classification groups (no AF, paroxysmal, persistent, permanent). (D) The probability of being in any of the device classification groups at any given AF burden level (conditional density plot). As AF burden increases, the probability of being classified in a progressively more severe classification (no AF, paroxysmal, persistent, permanent) increases. This is in contrast to the clinical AF classification (B).

(From Charitos EI, Pürerfellner H, Glotzer TV, et al. Clinical classifications of atrial fibrillation poorly reflect its temporal persistence: insights from 1,195 patients continuously monitored with implantable devices. J Am Coll Cardiol . 2014;63:2840–2848.)


AF can be classified as valvular or nonvalvular. The 2012 focused update of the European Society of Cardiology (ESC) guidelines defined valvular AF as rheumatic valvular disease (predominantly mitral stenosis) or prosthetic heart valves. Similarly, the 2014 American Heart Association/American College of Cardiology/Heart Rhythm Society (AHA/ACC/HRS) guidelines for the management of patients with AF defined nonvalvular AF as AF in the absence of rheumatic mitral stenosis or a mechanical heart valve, but explicitly added bioprosthetic heart valves or mitral valve repair within the “valvular heart disease” group.


The terms “lone” and “idiopathic” AF have been variably defined in the literature, but they generally refer to younger AF patients who have no clinical or echocardiographic evidence of cardiopulmonary disease, hypertension, or diabetes mellitus. However, this categorization is being abandoned since the category of lone AF no longer has mechanistic or clinical utility. Similarly, the term “chronic AF” has variable definitions and should not be used to describe populations of patients with AF.


Mechanism of Atrial Fibrillation


The pathogenesis of AF remains incompletely understood and is believed to be complex, multifactorial, and variable in different individuals. Two concepts of the underlying mechanism of AF have received considerable attention: factors that trigger AF and factors that perpetuate the arrhythmia. In general, patients with frequent, self-terminating episodes of AF are likely to have a predominance of factors that trigger AF, whereas patients with AF that does not terminate spontaneously are more likely to have a predominance of perpetuating factors. Although such gross generalization has clinical usefulness, often there is considerable overlap of these mechanisms. The typical patient with paroxysmal AF has identifiable ectopic foci initiating the arrhythmia, but these triggers cannot be recorded in all patients. Conversely, occasional patients with persistent or permanent AF can be cured of their arrhythmia by ablation of a single triggering focus, a finding suggesting that perpetual firing of the focus can potentially be the mechanism sustaining this arrhythmia in some cases.


Advanced mapping technologies, along with studies in animal models, have suggested the potential for complex pathophysiological substrates and modifiers responsible for AF ( Fig. 15.1 ), including the following: (1) continuous aging or degeneration of atrial tissue and the cardiac conduction system; (2) progression of structural heart disease (e.g., valvular heart disease and cardiomyopathy); (3) myocardial ischemia, local hypoxia, electrolyte derangement, and metabolic disorders (e.g., atherosclerotic heart disease, chronic lung disease, hypokalemia, and hyperthyroidism); (4) inflammation related to pericarditis or myocarditis, with or without cardiac surgery; (5) genetic predisposition; (6) drugs; and (7) autonomic influences.




Fig. 15.1


The Pathophysiological Triangle in Atrial Fibrillation (AF) .

(From Kottkamp H, Schreiber D. The substrate in “early persistent” atrial fibrillation. JACC Clin Electrophysiol . 2016;2:140–142.)


Mechanism of Initiation of Atrial Fibrillation


The factors responsible for the onset of AF include triggers that induce the arrhythmia and a receptive substrate that sustains it. The triggers are diverse yet may not cause AF in the absence of other contributors. There are two different types of arrhythmias that can potentially play a role in generating AF: premature atrial complexes (PACs) that initiate AF (focal triggers) and focal tachycardia that either induces fibrillation in the atria or mimics AF by creating a pattern of rapid and irregular depolarization wavefronts in the atria for as long as the focus continues to discharge.


The mechanism of initiation of AF is not certain in most cases and likely is multifactorial. Triggers propagating into the atrial myocardium can potentially initiate multiple reentrant wavelets and AF. In some patients with paroxysmal AF, impulses initiated by ectopic focal activity propagate into the left atrium (LA) and encounter heterogeneously recovered tissue. If reentry were assumed to be the mechanism of AF, initiation would require an area of conduction block and a wavelength of activation short enough to allow the reentrant circuits to persist in the myocardium.


Once triggered, AF can be self-sustained, in which case the continued firing of the focus may not be required for maintenance of the arrhythmia, and ablation of the focus may not terminate AF, but can potentially prevent the reinitiation of AF. Conversely, initiation and maintenance of AF can depend on uninterrupted periodic activity of a few discrete reentrant or triggered sources localized to the LA (i.e., focal drivers), emanating from such sources to propagate through both atria and interact with anatomic or functional obstacles, thus leading to fragmentation and multiple wavelet formation. Factors such as wavefront curvature, sink-source relationships, and spatial and temporal organization all are relevant to the understanding of the initiation of AF by the interaction of the propagating wavefronts with such anatomic or functional obstacles. Indeed, all these factors, which differ from triggers, importantly affect the initiators of AF.


AF triggering factors include sympathetic or parasympathetic stimulation, bradycardia, PACs (which may be the most common cause; Fig. 15.2 ), atrial flutter (AFL; see Fig. 12.2 ), supraventricular tachycardias (SVTs; especially those mediated by atrioventricular [AV] bypass tracts [BTs]; Fig. 15.3 ), and acute atrial stretch. Identification of these triggers has clinical importance because treatment approaches directed at elimination of the triggers (e.g., catheter ablation of the initiating PACs or SVT) can be curative in selected patients.




Fig. 15.2


Atrial Fibrillation (AF) Induction by Premature Atrial Complexes (PACs) Originating From the Right Superior Pulmonary Vein (PV) .

Two monomorphic PACs (arrows) occur at short coupling intervals and are inscribed within the T wave. The second PAC (red arrows) triggers AF.



Fig. 15.3


Atrial Fibrillation (AF) Induction by Orthodromic Atrioventricular Reentrant Tachycardia (AVRT) .

CS dist , Distal coronary sinus; CS prox , proximal coronary sinus; HRA, high right atrium; RVA , right ventricular apex.


PV triggers.


Triggering foci of rapidly firing cells within the sleeves of atrial myocytes extending into the pulmonary veins (PVs) have been shown to be the underlying mechanism in most cases of paroxysmal AF. Supporting this idea are clinical studies of impulses generated by single foci propagating from individual PVs or other atrial regions to the remainder of the atria as fibrillatory waves and abolition of AF by radiofrequency (RF) ablation to eliminate or isolate the PV foci. The PVs also represent the main trigger site for AF initiation in patients with recurrent persistent AF, with an overall prevalence similar to that found in patients with paroxysmal AF.


Based on several features, the thoracic veins are highly arrhythmogenic. The PV-LA junction has discontinuous myocardial fibers separated by fibrotic tissues and hence is highly anisotropic. Insulated muscle fibers can promote reentrant excitation, automaticity, and triggered activity. These regions likely resemble the juxtaposed islets of atrial myocardium and vascular smooth muscle in the coronary sinus (CS) and AV valves that, under normal circumstances, manifest synchronous electrical activity but develop delayed after-depolarizations and triggered activity in response to catecholamine stimulation, rapid atrial pacing, or acute stretch.


Furthermore, the PVs of patients with paroxysmal AF demonstrate abnormal properties of conduction so that there can be markedly reduced refractoriness within the PVs, progressive conduction delay within the PV in response to rapid pacing or programmed stimulation, and often conduction block between the PV and the LA. Such findings are much more common in patients with paroxysmal AF than in normal subjects. Rapidly firing foci can often be recorded within the PVs with conduction block to the LA. Administration of catecholamines such as isoproterenol can lead to shortening of the LA refractory period, thereby allowing these foci to propagate to the LA and induce AF. These discontinuous properties of conduction within the PV can also provide a substrate for reentry within the PV itself, although this remains to be proven.


Non-PV triggers.


Although more than 90% of AF triggering foci that are mapped during electrophysiological (EP) studies in patients with paroxysmal AF occur in the PVs, foci within the superior vena cava (SVC), small muscle bundles in the ligament of Marshall, and the musculature of the CS have been identified. Although these latter locations of triggering foci are uncommon in patients with paroxysmal AF, the common factor is that the site of origin is often within a venous structure that connects to the atrium. Other sites of initiating foci can be recorded in the LA wall or along the crista terminalis in the right atrium (RA).


Mechanism of Maintenance of Atrial Fibrillation


Having been initiated, AF can be brief; however, various factors can act as perpetuators, thus ensuring the maintenance of AF. One factor is the persistence of the triggers and initiators that induced the AF, which then act as an engine driving the continuation of AF. In this setting, maintenance of AF is dependent on the continued firing of the focus (the so-called “focal driver”). Alternatively, AF can persist even in the absence of the focal drivers. Without focal drivers, persistence of AF results from a combination of electrical and structural remodeling processes characterized by atrial dilation and shortening of atrial refractoriness (see later). These factors can be present at baseline but can also be induced by the AF itself.


The mechanisms responsible human AF remain controversial. Several theories have been proposed to explain the EP mechanisms underlying AF, including the multiple wavelet theory and the localized source hypothesis.


Multiple wave reentry hypothesis.


For many years, the “multiple wave reentry” hypothesis was the most widely held theory on the maintenance of AF, and was a key development in our understanding of the mechanism of AF. On the basis of a computer model of AF, Moe and associates hypothesized that AF is sustained by multiple randomly wandering wavelets in both atria that collide with each other and extinguish themselves or create new, daughter wavelets that continually reexcite the atria and perpetuate the arrhythmia. Those functional reentrant circuits are therefore unstable; some disappear, whereas others reform. These circuits have variable, but short, cycle lengths (CLs) to which atrial tissue cannot respond in a 1 : 1 fashion. As a result, functional block, slow conduction, and multiple wavefronts develop. It has been suggested that at least four to six independent wavelets are required to maintain AF. These wavelets rarely reenter themselves but can reexcite portions of the myocardium recently activated by another wavefront, a process called random reentry. In simulated cardiac tissue, multiple wavelet fibrillation is equivalent to spiral/scroll waves that are inherently unstable and spontaneously develop wavebreaks along the arm of the rotor which then form daughter wavelets. Because multiple wave reentry fibrillation is purely reentrant, its initiation requires a trigger to create the original unstable spiral/scroll wave that subsequently breaks up to create daughter wavelets; however, once initiated, additional triggers are no longer required to maintain fibrillation.


The persistence of multiple-circuit reentry depends on the ability of a tissue to maintain enough simultaneously reentering wavefronts so that electrical activity is unlikely to extinguish simultaneously in all parts of the atria. Therefore the larger the number of wavelets present, the more likely the arrhythmia will be sustained. The number of wavelets coexisting at any moment depends on the atrial mass, excitation wavelength, refractory period, conduction velocity, and anatomic obstacles in different portions of the atria. In essence, a large atrial mass with short refractory periods and prominent conduction delay would yield increased wavelets and would present the most favorable situation for AF to be sustained.


Clinical support for this hypothesis seemed to come from the surgical maze and some substrate-based catheter ablation procedures, which were proposed to result in dividing the atrial into small electrical compartments and, thus, disallowing maintenance of the randomly propagating wavelets. The reentrant circuits that comprise multiple wave reentry are functional, multiple, and dynamic; thus ablation of multiple wave reentry is not aimed at eliminating the possibility of its existence but at maximizing the probability of its spontaneous termination through collisions between circuit cores and unexcitable tissue boundaries via atrial debulking and compartmentalization.


Localized source hypothesis.


In contrast to the nonhierarchical, self-sustaining disorganized electrical activity implicated in the multiple wavelet theory, recent evidence suggests the presence of hierarchical electrical organization in which localized sources drive disorganized activity. This hypothesis suggests that AF is intermittently maintained by a small number of localized (spatially stable) high-frequency sources with periods of self-sustaining disorganization. Rotors and focal sources exhibit 1 : 1 activation within their spatial domain, with peripheral disorganization. This concept was supported by experimental studies using high-resolution optical mapping, which demonstrated spatial and temporal organization during AF. Localized sources can be either discrete foci with centrifugal spread of activation or small anatomic reentry circuits or functional rotors.


When cardiac impulses are continuously generated at a rapid rate from any source or any mechanism, they activate the tissue of that cardiac chamber in a 1 : 1 manner, up to a critical rate. Once this critical rate is exceeded, not all the tissue of that cardiac chamber can respond in a 1 : 1 fashion (e.g., because the CL of the driver is shorter than the refractory periods of those tissues), and “fibrillatory conduction” develops. Fibrillatory conduction can be caused by spatially varying refractory periods or by the structural properties of atrial tissue, with source-sink mismatches providing spatial gradients in the response. Fibrillatory conduction is characterized by activation of tissues at variable CLs, all longer than the CL of the driver, because of variable conduction block; in that manner, activation is fragmented. This is the mechanism of AF in several animal models in which the driver consists of a stable, abnormal automatic focus of a very short CL, a stable reentrant circuit with a very short CL, or an unstable reentrant circuit with a very short CL. It also appears to be the mechanism of AF in patients in whom activation of the atria at very short CLs originates in one or more PVs. The impulses from the PVs seem to precipitate and maintain AF.


The concept of fibrillatory conduction is relevant to the “mother rotor fibrillation” hypothesis, in which a fast stationary or meandering spiral/scroll wave in one region of the tissue develops peripheral wavebreaks as the spiral/scroll arm propagates into surrounding tissue with longer refractory periods. Though some investigators suggest that these mother rotors are likely fixed, others have suggested that they may precess (i.e., wobble), albeit in small fairly well-defined areas. In atria with extensive fibrosis, multiple stable rotors can possibly coexist in different regions, insulated by intervening tissue that cannot maintain 1 : 1 conduction. This variant is equivalent to mother rotor fibrillation with multiple stable mother rotors. Unlike multiple wavelet fibrillation in which the functional reentry is inherently unstable and nonlocalized and the spontaneous peripheral wavebreaks play a causal role in both initiating and maintaining fibrillation, “mother rotor fibrillation” is driven by a localized source and the peripheral wave breaks are noncausal epiphenomena. However, similar to multiple wave reentry fibrillation, mother rotor fibrillation is purely reentrant and requires a trigger to initiate the original rotor; once initiated, no further triggers are necessary to perpetuate fibrillation.


Recently, several mapping studies have provided clinical evidence of the localized source hypothesis by demonstrating that rotational or focal drivers in localized regions maintain AF, and that AF could be eliminated by directly ablating sites of rotors and focal sources that exhibit high-frequency, periodic activity, based on either electrogram visual analysis, dominant frequency analysis, or panoramic endocardial mapping using phase analysis (see below).


Mapping human AF.


The complexity of AF electrograms, with varying amplitude and morphology as well as spatiotemporal CL variations, poses significant challenges to mapping human AF. Recent clinical studies have demonstrated conflicting mechanisms underlying sustained AF; one source of the discrepancies likely arises from differences in scale (global vs. regional) and spatial resolution of the mapping techniques. A second possible factor is the approach employed to analyze fibrillatory wavefront dynamics, using either phase mapping or activation time.


Conventional activation mapping techniques used in the EP lab, which assign a specific time point on the unipolar electrogram as the marker of local activation, cannot be employed during AF; fibrillation electrograms typically are fractionated and low amplitude, and are prone to artifacts. Spectral analysis and phase mapping of cardiac potentials have been used by researchers to overcome some of those challenges. Both algorithms derive originally from optical mapping experiments conducted in ex vivo hearts. Although optical mapping provides much higher spatiotemporal resolution and accuracy when tracking rotor formation and maintenance, current optical mapping strategies do not allow panoramic endocardial mapping and require the use of potentially toxic voltage-sensitive dyes that preclude their use for human in vivo studies.


The “spectrum” of a signal displays its energy distribution in the frequency domain. Frequency domain analysis can identify the frequency of the highest peak in the spectrum (i.e., “dominant frequency”), which is often used as a surrogate for the average activation rate at a specific location. Hence, spectral analysis can map the distribution of AF frequencies and localize the areas with the highest activation frequencies (i.e., the shortest CLs, dominant frequency), which usually coincide with the location of AF sources (foci or rotors) that maintain AF. Those techniques have demonstrated that AF has significant periodic elements with varying degrees of regularity, and that certain regions of the atria can have higher activation frequencies than other regions, suggesting that these areas can potentially be the drivers that maintain AF and can provide targets of ablation therapy. A limitation of spectral analysis is that frequency domain mapping produces time-averaged frequency information over space, thereby losing the ability to track temporal variations of the signal.


“Phase” is a measure of where a signal is in its cycle of oscillation at a given point in time. Phase mapping computes oscillations in the signal over its entire duration, independent of its amplitude, and characterizes a temporal signal in its instantaneous phase of the activation/recovery cycle at each time point. Computation of phase involves the transformation of the signal to the phase domain by selecting a specific CL from the temporal signal. Analysis of phase changes over space provides information on the patterns of organization (repetitive activity) and enables visualization of the spatiotemporally distributed patterns of propagation during cardiac fibrillation. Importantly, phase mapping makes it possible to detect rotational wavefronts (rotors) and determines their center of rotation (singularity point) ( eFig. 15.2 ). However, phase mapping has the propensity to introduce false rotors during complex activation patterns, such as wavefront propagation about a line of block.





eFig. 15.2


Varying Activation Patterns Identified in Phase Movies From the Left Atrium of the Sheep Heart.

(A) Sequential snapshots show a rotor pivoting around a phase singularity (point where all phases converge). (B) Breakthrough activation pattern. The wave seems on the center of the field of view and propagates outward. Right, Key for the different phases of the action potential is color-coded.

(From Quintanilla JG, Pérez-Villacastín J, Pérez-Castellano N, et al. Mechanistic approaches to detect, target, and ablate the drivers of atrial fibrillation. Circ Arrhythmia Electrophysiol . 2016;9:1–12.)


In the clinical setting, recent studies employing panoramic atrial mapping during AF have used phase analysis of contact atrial endocardial electrograms from basket electrode catheters (focal impulse and rotor modulation [FIRM], Fig. 15.4 ) or reconstructed atrial electrograms from body surface recordings (electrocardiogram [ECG] imaging, eFig. 15.3 ). Those studies demonstrated the presence of atrial driver regions with a high prevalence of transient rotors, and suggested that AF is sustained by spiral waves (rotors) or focal sources, or both, which are sufficiently stable in space to be targeted by ablation. Both approaches identified phase singularity points (core of a rotor) as a target for ablation. Elimination of those rotors and focal source could acutely modify or terminate AF and substantially improve ablation outcome. Localized sources can potentially explain modulation of AF by ablation insufficient to limit critical mass and AF termination by localized intervention.




Fig. 15.4


Focal Sources of Atrial Fibrillation Identified by Focal Impulse and Rotor Modulation (FIRM) Mapping.

(A) Fluoroscopic view (left anterior oblique) of the multielectrode basket catheter deployed in the left atrium. (B) FIRM mapping shows a left atrial rotor with counterclockwise rotation (from red to blue on the activation time scale with red representing early activation). (C) FIRM mapping shows a left atrial focal impulse originating from electrodes 2 and 3 spline C and D of the basket catheter (red represents early activation with a centrifugal pattern).

(From Sommer P, Kircher S, Rolf S, et al. Successful repeat catheter ablation of recurrent longstanding persistent atrial fibrillation with rotor elimination as the procedural endpoint: a case series. J Cardiovasc Electrophysiol . 2016;27:274–280.)





eFig. 15.3


Phase Mapping of Electrocardiogram Imaging Data Showing Posterior View of Left Atrium During Paroxysmal Atrial Fibrillation.

Panel A shows serial snapshots of a single wave emerging out of the left inferior pulmonary vein (PV, white star ) and reaching right PVs in 30 milliseconds while it expands radially to the roof and inferior walls. Panel B shows serial snapshots of two successive rotations (white arrows) of a rotor located near the ostia of right PVs. The core of the rotor ( white star at the center of rainbow-colored phases of rotor) is seen meandering in a small region in this example. The blue wave indicates the depolarizing front, which makes one full rotation in 160 milliseconds. The phases of wave propagation are color coded using rainbow scale. The blue color represents depolarizing wave and the green represents the end of repolarization. The wavefront can be read by following the blue color. The time (milliseconds) at the bottom of each snapshot represents the moment in the time window when the snapshot was taken. LIPV, Left inferior PV; LSPV, left superior pulmonary vein; RIPV, right inferior PV; RSPV, right superior PV; SVC, superior vena cava.

(From Haissaguerre M, Hocini M, Shah AJ, et al. Noninvasive panoramic mapping of human atrial fibrillation mechanisms: a feasibility report. J Cardiovasc Electrophysiol . 2013;24:711–717.)


It is important to note that while both ECG imaging and FIRM mapping use phase analysis to determine rotational conduction behavior during AF, detection of rotors using those techniques provides fundamentally different results regarding the spatial behavior of rotors. FIRM mapping demonstrates spatially stable rotors in both atria; many of these rotors are detectable for many seconds up to minutes at the same location. In contrast, ECG imaging techniques show that the reentrant circuits are not spatially stable but meandered substantially throughout the atrium; nevertheless, they recurred repetitively at specific sites that can potentially provide a target for ablation. The reason for the significant discrepancy between the spatial behavior of rotors identified by these different techniques is currently unclear. In a recent study comparing resolution requirements for detection of stationary and meandering rotors found that rotor trajectories can be lost at resolutions for which stable rotors are still identifiable, which may explain differences in findings on rotor stability with basket catheters identifying stable rotors and noninvasive ECG imaging identifying transient meandering rotors.


However, despite these data, the rotor paradigm is neither confirmed nor universally accepted, with recent studies raising questions about the efficacy of rotor-targeted ablation. Sufficient spatial resolution is essential for the accurate detection of rotors and focal sources. Although stationary rotors may be identified at coarse resolutions, meandering rotors are lost. While most clinically used catheters offer sufficient spatial resolution to identify and track rotor core location for the range of wavelengths occurring in human AF, mapping catheters with localized coverage (e.g., quadripolar or ring catheters) are not capable of tracking rotors unless the catheter is fortuitously placed over a rotor that does not meander outside the margins of the catheter poles. In contrast, basket catheters provide global coverage. However, the low resolution of the basket catheters renders them prone to false detections; interpolation of phases is inherently biased toward detection of rotors as the algorithm is devised to demonstrate rotational activity, and focal activation might be displayed as rotational activity if the wavefront reaches the surrounding electrodes sequentially. This can potentially explain, in part, the large incidence of rotors using FIRM mapping, the low AF termination rate with ablation, and poor long-term success for ablating rotors detected by basket catheters. On the other hand, ECG imaging lacks the ability to discern among epicardial breakthroughs, spontaneous depolarizations, and (subcentimeter) microreentry, which are seen as focal activity.


Substrate for Atrial Fibrillation


As noted, AF results from the interplay between a trigger for initiation and a vulnerable EP substrate for maintenance. The fact that most potential triggers do not initiate AF suggests some role for functional and structural substrates in most patients. However, the relative contribution of triggers versus substrate can vary with the clinical context, and the exact nature of the interaction between triggers and substrate remains to be elucidated.


AF commonly occurs in the context of other cardiac or noncardiac pathological conditions, such as valvular disease, hypertension, ischemic heart disease, heart failure, or hyperthyroidism. Depending on the type, extent, and duration of such external stressors, a cascade of time-dependent adaptive, as well as maladaptive, atrial responses develops to maintain homeostasis (so-called “atrial remodeling”), including changes at the ionic channel level, cellular level, extracellular matrix level, or a combination of these, which result in electrical, functional, and structural consequences.


A hallmark of atrial structural remodeling is atrial dilation, often accompanied by a progressive increase in interstitial fibrosis. Atrial arrhythmias, especially AF, are the most common manifestations of electrical remodeling. Increased dispersion in atrial refractoriness and inhomogeneous dispersion of conduction abnormalities, including block, slow conduction, and dissociation of neighboring atrial muscle bundles, are key elements in the development of the substrate of AF. Importantly, different pathological conditions can be associated with a different set of remodeling responses in the atria.


Even in the setting of the AF occurring in the absence of apparent structural heart disease, there is accumulating evidence that occult abnormalities (e.g., patchy fibrosis, inflammatory infiltrates, loss of myocardial voltage, conduction slowing, altered sinus node function, and vascular dysfunction) can be observed and likely represent an early stage of atrial remodeling contributing to the substrate of AF.


Atrial Electrophysiological Properties


In normal atrial cardiomyocytes, phase 0 of the action potential is mediated by the rapidly activating sodium (Na + ) current (INa). These potentials are called fast response potentials ( see Chapter 1 ). As a result, the atrium has several properties that permit the development of very complex patterns of conduction and an extremely rapid atrial rate, as seen in AF. The action potential duration is relatively short, and reactivation can occur partially during phase 3 and usually completely within 10 to 50 milliseconds after return to the diastolic potential. The refractory period shortens with increasing rate, and very rapid conduction can occur.


Patients with AF and no apparent structural heart disease appear to have increased dispersion of atrial refractoriness, which correlates with enhanced inducibility of AF and spontaneous episodes. Some patients have site-specific dispersion of atrial refractoriness and intraatrial conduction delays resulting from nonuniform atrial anisotropy.


Atrial Fibrosis


Atrial fibrosis plays an important role in the pathophysiology of AF. Atrial fibrosis results from various cardiac insults that share common fibro-proliferative signaling pathways. Fibrotic myocardium exhibits slow and inhomogeneous conduction, with spatial “nonuniform anisotropic” impulse propagation, likely secondary to reduced intercellular coupling, discontinuous branching architecture, and zigzagging circuits. When combined with inhomogeneous dispersion of refractoriness within the atria, conduction block provides milieu necessary for the development of reentry. The greater the slowing of conduction velocity in fibrotic myocardium, the shorter the anatomic circuit needed to sustain a reentrant wavelet. In fact, reentrant circuits need be only a few millimeters in length in discontinuously conducting tissue. Thus atrial regions with advanced fibrosis can harbor local sources for AF. Such a hypothesis would not preclude the remainder of the atria from showing fibrillatory conduction or functional reentrant waves.


Increased atrial fibrosis is a manifestation of the normal aging process as well as various pathological conditions such as hypertension, coronary artery disease, heart failure, and sleep apnea. Increased atrial fibrosis has been demonstrated even in patients with so-called “lone” or “idiopathic” AF, suggesting that AF in these patients potentially represents an arrhythmic manifestation of “fibrotic atrial myopathy.” The strong association of sinus node dysfunction (SND) and AF (the bradycardia-tachycardia syndrome) also suggests that replacement of atrial myocytes by fibrosis likely plays an important part in the pathogenesis of AF, although in some instances the bradycardia component is a functional response to the tachycardia. Recent studies using delayed-enhancement cardiovascular magnetic resonance (CMR) imaging and electroanatomic voltage mapping demonstrated that the extent and distribution of the fibrotic atrial changes can vary widely among patients with AF. Nonetheless, a higher degree of fibrosis often is observed in patients with persistent AF versus paroxysmal AF. In addition, atrial fibrosis defined by delayed-enhancement CMR was found to be independently associated with AF recurrence in patients undergoing catheter ablation procedures.


Importantly, AF itself seems to produce various alterations of atrial architecture that further contribute to atrial remodeling, mechanical dysfunction, and perpetuation of fibrillation. Longstanding AF results in loss of myofibrils, accumulation of glycogen granules, disruption in cell-to-cell coupling at gap junctions, and organelle aggregates.


Changes in AF characteristics during evolving fibrosis also have a direct impact on why electrical or drug treatment ultimately fails to achieve conversion to NSR. In the markedly fibrotic and discontinuous atrial tissue, characterized by discontinuous anisotropy, marked degree of gap junctional uncoupling, and fiber branching, the safety factor for propagation is higher than in normal tissue. Hence, blocking of the Na + current to the same degree as is necessary for the termination of functional reentry may not terminate reentry caused by slow and fractionated conduction in fibrotic scars of remodeled atria. Conduction in discontinuous tissue is mostly structurally determined and leads to excitable gaps behind the wavefronts. If a gap is of critical size, the effectiveness of drugs that prolong atrial refractoriness will be limited. Furthermore, scar tissue is likely to exhibit multiple entry and exit points and multiple sites at which unidirectional block occurs. This can potentially lead to activity whose appearance in local extracellular electrograms changes from beat to beat, as well as beat-to-beat CL variability. Although such regions can be expected to respond to defibrillation, AF can resume after extrasystoles or normal sinus beats immediately after conversion, with unidirectional block recurring because of scar.


Atrial Stretch


Atrial stretch and dilation can play a role in the development and persistence of AF. Clinically, AF episodes occur more frequently in association with conditions known to cause elevated LA pressure and atrial stretch, such as acutely decompensated systolic or diastolic heart failure. In addition, the echocardiographic LA volume index and restrictive transmitral Doppler flow patterns are strong predictors of the development of AF.


The structure of the dilated atria can potentially have important EP effects related to stretch of the atrial myocardium (so-called “electromechanical feedback”). Acute atrial stretch reduces the atrial refractory period and action potential duration and depresses atrial conduction velocity, potentially through a reduction of cellular excitability by the opening of stretch-activated channels or changes in cable properties (membrane resistance, capacitance, core resistance). Regional stretch for less than 30 minutes activates the immediate early gene program, thus initiating hypertrophy and altering action potential duration in affected areas. Moreover, acutely altered stress and strain patterns augment the synthesis of angiotensin II, which induces myocyte hypertrophy. Angiotensin II can contribute to arrhythmogenic electrical dispersion by regionally increasing L-type calcium (Ca 2+ ) current (ICaL) and decreasing the transient outward potassium (K + ) current (Ito). Altered stretch of atrial myocytes also results in opening of stretch-activated channels, increasing G protein–coupled pathways. This leads to increased protein kinase A and C activity, and enhanced ICaL through the cell membrane, and increased release of Ca 2+ from the sarcoplasmic reticulum, thus promoting afterdepolarizations and triggered activity. Furthermore, acute stretch can promote an increase in dispersion of refractoriness and spatial heterogeneity by causing conduction block and potentially contributing to the development of AF. These alterations occur nonuniformly because stretch is greater in areas of thin versus thick atrial myocardium.


In addition, chronic atrial stretch, as a result of AF and several conditions associated with AF, can promote atrial fibrosis via the activation of multiple profibrotic and hypertrophic signaling pathways.


Inflammation


There is increasing evidence that implicates inflammation (and its downstream effects, including atrial fibrosis) in the pathogenesis of AF. Clinically, AF occurs frequently in the setting of inflammatory states such as cardiac surgery and acute pericarditis. In addition, the levels of inflammatory biomarkers (C-reactive protein [CRP] and interleukin-6 [IL-6]) are significantly increased in patients with AF, findings suggesting the presence of systemic inflammation in these patients. Elevation of the levels of CRP and IL-6 has been shown to predict future development, recurrence, and burden of AF. There is also evidence suggesting that inflammation is involved in electrical and structural atrial remodeling. Furthermore, inflammation appears to increase the inhomogeneity of atrial conduction directly, potentially via disruption of expression of connexin proteins, leading to impaired intercellular coupling.


It is also likely that inflammation can be a consequence of AF. CRP levels decrease following restoration of NSR. Rapid atrial activation in AF results in Ca 2+ overload in atrial myocytes that can potentially result in cell death, which induces a low-grade inflammatory response. The inflammation, in turn, can induce healing and reparative fibrosis that likely enhance remodeling and promote perpetuation of the arrhythmia.


Currently, the exact role of inflammation in AF is poorly defined, and it remains unclear whether inflammation is actually involved in the mechanisms underlying AF or whether it is simply an epiphenomenon. Although therapies directed at attenuating the inflammatory burden (e.g., glucocorticoids, statins, and angiotensin II inhibitors) appear promising, early clinical trials do not support a significant benefit.


Atrial Remodeling in Atrial Fibrillation


AF is a progressive arrhythmia. In 14% to 24% of patients with paroxysmal AF, persistent AF develops, even in the absence of progressive underlying heart disease. Furthermore, conversion of AF to NSR, electrically or pharmacologically, becomes more difficult when the arrhythmia has been present for a longer period. In fact, the arrhythmia itself results in a cascade of electrical and structural changes in the atria that are themselves conducive to the perpetuation of the arrhythmia (“AF begets AF”), a process known as remodeling. Recurrent AF can potentially lead to irreversible atrial remodeling and eventually permanent structural changes that account for the progression of paroxysmal to persistent and finally to permanent AF, characterized by the failure of electrical cardioversion and pharmacological therapy to restore and maintain NSR. Even after cessation of AF, these abnormalities persist for periods that vary in proportion to the duration of the arrhythmia.


Changes in atrial EP features induced by AF can occur through alterations in ion channel activities that cause partial depolarization and abbreviation of atrial refractoriness. These changes promote the initiation and perpetuation of AF (electrical remodeling) and the modification of cellular Ca 2+ handling, which causes contractile dysfunction (contractile remodeling), as well as atrial dilation with associated structural changes (structural remodeling). Experimentally, electrical and contractile remodeling begins shortly after the onset of AF, with a parallel decrease in both atrial refractory period and contractility over the first minutes of AF. This is followed by further abbreviation in atrial refractoriness and increase in atrial dimensions over the following days. Structural changes follow a much slower time course, likely starting after several weeks.


Electrical Remodeling


Electrical remodeling results from the high rate of electrical activation. The EP changes typical of atrial myocytes during AF include shortening of the action potential duration and atrial refractory period, and reduction in the amplitude of the action potential plateau. Furthermore, AF results in deficiency in the ability of the repolarization time course (action potential duration) to adapt to changes in rate (“abnormal restitution”); consequently, the atrial refractory period fails to lengthen appropriately at slow rates (e.g., with return to NSR). These changes can contribute to the stability of a longer lasting form of AF because, according to the multiple wavelet theory, a short wavelength results in smaller wavelets, which increase the maximum number of wavelets, given a certain atrial mass. Tachycardia-induced changes in refractoriness are spatially heterogeneous, and there is increased variability both within and among various atrial regions, which can promote atrial vulnerability and AF maintenance and provide a substrate for reentry.


The mechanisms for electrical remodeling and shortening of the atrial refractory period are not entirely clear. Several potential explanations exist, including ion channel remodeling, angiotensin II, and atrial ischemia. The principal components of electrical remodeling include reduction in the L-type Ca 2+ current (ICaL), rectifier background K + current (IK1), and constitutive acetylcholine-regulated K + current (IKACh), and abnormal expression and distribution of the gap junctions.


Downregulation of ICaL seems to be responsible for shortening of the atrial action potential, whereas a decrease in Ito is considered to result in loss of physiological rate adaptation of the action potential. The fast atrial rate during AF causes accumulation of intracellular Ca 2+ . The reduction in ICaL can be explained by a decreased expression of the L-type Ca 2+ channel α1C subunit, likely as a compensatory mechanism to minimize the potential for cytosolic Ca 2+ overload secondary to increased Ca 2+ influx during the rapidly repetitive action potentials during AF. Verapamil, an L-type Ca 2+ channel blocker, was shown to attenuate electrical remodeling and hasten complete recovery without affecting inducibility of AF, whereas intracellular Ca 2+ overload, induced by hypercalcemia or digoxin, enhances electrical remodeling. Electrical remodeling can be attenuated by the sarcoplasmic reticulum’s release of the Ca 2+ antagonist ryanodine, a finding suggesting the importance of increased intracellular Ca 2+ to the maladaptation of the atrial myocardium during AF.


Angiotensin II may also be involved in electrical remodeling, and angiotensin II inhibitors may prevent the remodeling process. Angiotensin-converting enzyme inhibitors reduce the incidence of AF in patients with left ventricle (LV) dysfunction after myocardial infarction (MI) and in patients with chronic ischemic cardiomyopathy. Atrial ischemia is another possible contributor to electrical remodeling and shortening of the atrial refractory period via activation of the Na + -H + exchanger.


More recently, microRNAs (miRNAs or miRs), a group of small noncoding RNA molecules that negatively regulate gene expression, were found to have an important role in a wide range of electrical and structural atrial remodeling processes. Furthermore, experimental studies showed that the rapid rates of AF can lead to autonomic atrial remodeling, with heterogeneous increase in atrial sympathetic innervation, which can potentially promote enhanced automaticity, triggered activity, and spatially heterogeneous abbreviation of refractoriness.


Furthermore, persistent AF can result in other changes within the atria, including gap junctional remodeling, manifest as an increase in the expression and distribution of connexin 43 and heterogeneity in the distribution of connexin 40, both of which are intercellular gap junction proteins.


Contractile and Structural Remodeling


Sustained AF has also been associated with structural changes such as myocyte hypertrophy, myocyte death, tissue fibrosis, impaired atrial contractility, and atrial stretch and dilation. Atrial dilation increases electrical instability by shortening the effective refractory period and slowing atrial conduction. These structural changes, many of which probably are irreversible, appear to occur over periods of weeks to months.


Cellular remodeling is caused by the apoptotic death of myocytes with myolysis. AF results in marked changes in atrial cellular substructures, including loss of myofibrils, accumulation of glycogen, changes in mitochondrial shape and size, fragmentation of sarcoplasmic reticulum, and dispersion of nuclear chromatin.


Contractile remodeling is likely caused by downregulation of ICaL (resulting in reduced release of Ca 2+ during systole), as well as myolysis (loss of sarcomeres). Contractile remodeling can potentially cause thrombus formation and atrial dilation. Contractile remodeling starts early after onset of AF, and its recovery generally takes longer than reversal of electrical remodeling, likely because of the time it takes for the atria to replace lost sarcomeres.


In addition to remodeling of the atria, the sinus node can undergo remodeling, resulting in SND and bradyarrhythmias caused by reduced sinus node automaticity or prolonged sinoatrial conduction. The phenomenon of sinus node remodeling likely contributes to the episodes of bradycardia seen in the tachycardia-bradycardia syndrome and may reduce sinus rhythm stability and increase the stability of AF. As mentioned earlier, elements of the sinus bradycardia appear to be functionally reversible if the tachycardia is prevented.


Studies suggest that the PVs are more susceptible to electrical alterations resulting from AF than the atria. Although the PVs display significantly longer refractory periods at baseline than the atria, they exhibit more prominent shortening of refractoriness after a brief episode of pacing-induced AF. Moreover, the short-term presence of AF does influence PV EP properties by slowing the conduction velocity without affecting the conduction times of the atria. Structural changes in the atria after remodeling, such as stretch, can also lead to increased PV activity. Atrial stretch can lead to increased intraatrial pressure, causing a rise in the rate and spatiotemporal organization of electrical waves originating in the PVs. These changes imply that electrical and structural remodeling increases the likelihood of ectopic PV automaticity and AF maintenance.


Tachycardia-induced atrial remodeling can potentially underlie various clinically important phenomena, such as the tendency of patients with other forms of supraventricular arrhythmias to develop AF, the tendency of AF to recur early after electrical cardioversion, the resistance of longer duration AF to antiarrhythmic medications, and the tendency of paroxysmal AF to become persistent.


If NSR is restored within a reasonable time period, EP changes and atrial electrical remodeling appear to normalize gradually, atrial size decreases, and atrial mechanical function is restored. These observations lend support to the idea that the negative downhill spiral in which AF begets AF can be arrested with NSR that perpetuates NSR, and restoration of NSR may forestall progressive remodeling and the increase in duration and frequency of arrhythmic episodes by reverse remodeling.


Role of Autonomic Nervous System in Atrial Fibrillation


Cardiac function is modulated by both the extrinsic and the intrinsic cardiac autonomic nervous systems. The extrinsic (central) system is composed of sympathetic and parasympathetic components, and includes neurons in the brain and spinal cord and nerves directed to the heart. The intrinsic system is composed of a large network of autonomic cardiac ganglia buried throughout the epicardial fat within the pericardial space and in the ligament of Marshall. Groups of several cardiac ganglia comprise plexuses that coalesce in specific locations, and different groups of ganglia have different sites of innervation throughout the heart. Atrial ganglia contain afferent neurons from the atrial myocardium and from the central autonomic nervous system, and efferent cholinergic and adrenergic neurons, with heavy innervation of the PV myocardium and the atrial myocardium surrounding the ganglionic plexuses. In addition, an extensive array of interconnecting neurons creates a communication network among the different ganglionic plexuses, as well as between the ganglionic plexuses and the atrium and PV myocardium. The intrinsic system receives input from the extrinsic system and but acts independently to modulate numerous cardiac functions, including automaticity, contractility, and conduction.


Several studies have demonstrated that both divisions of the autonomic nervous system are involved in the initiation, maintenance, and termination of AF, with a predominant role of the parasympathetic system. Enhanced sympathetic activity shortens the atrial refractory period and also increases sarcoplasmic reticulum calcium release in the atrium and PV myocardium, promoting after-depolarization–related PACs and atrial tachycardia (AT), which in turn can initiate AF. On the other hand, increased vagal tone is frequently involved in the onset of AF in patients with structurally normal hearts. Parasympathetic stimulation results in nonuniform shortening of the atrial effective refractory periods, thereby setting up substrate for reentry, promoting the initiation and perpetuation of AF. Vagal stimulation can also lead to the emergence of focal triggers in the atrium. It is important to note that imbalances in the intrinsic cardiac nervous system (rather than an enhanced tone per se) are thought to be involved in the pathogenesis of AF. A shift toward an increase in sympathetic tone or toward a loss of vagal tone has been observed before postoperative paroxysmal AF, whereas a shift toward vagal predominance was observed in young patients with nocturnal episodes of paroxysmal AF.


The electrical properties of the PVs also are modulated by changes in autonomic tone. Anatomic studies revealed that the PVs and the adjoining posterior LA have a unique autonomic profile that differs from the rest of the atria, which likely contributes to the genesis of both focal triggers and sustained microreentry in this region. Ganglionated plexuses cluster within fat pads at the PV entrances and heavily innervate each of the myocardial sleeves of the four PVs. Activation of the ganglionic plexuses at the PV-LA junction promotes AF by a combined parasympathetic and sympathetic action.


Studies suggest that the extrinsic autonomic input to the heart (i.e., from the brain and spinal cord) exerts inhibitory control over the ganglionated plexuses and that attenuation or loss of this control would allow the ganglionated plexuses to become hyperactive. The ganglionic plexuses function as “integration centers” that modulate autonomic innervation. Hyperactivity of ganglionic plexuses can be proarrhythmic while low-level activity may be antiarrhythmic. Recent evidence suggests that noninvasive cardiac autonomic neuromodulation therapies (such as transcutaneous electrical stimulation of the vagus nerve) suppress ganglionated plexus activity, with consequent increased effective refractory period of the atrial and PV myocardium and suppressed AF inducibility. Other approaches to modulation of the extrinsic cardiac autonomic nervous system (e.g., renal sympathetic denervation and ganglion stellatum ablation) are being investigated for antiarrhythmic treatment. Furthermore, ablation of ganglionated plexuses located at the atrial entrances or antra of the PVs can potentially abolish or reduce AF inducibility.


Role of the Pulmonary Veins in Atrial Fibrillation


There is little controversy now that the PVs play a major role in triggering and maintaining AF, as established by animal and human models, especially in the setting of paroxysmal AF. Fibrillatory conduction is likely initiated by rapid discharges from one or several focal sources within the atria; in most patients with AF (94%), the focus is in one of the PVs (see Fig. 15.2 ). Extra-PV sites can also trigger AF, but this occurs in a minority of cases, likely no more than 10% of patients. AF is also perpetuated by microreentrant circuits, or rotors, that exhibit high-frequency periodic activity from which spiral wavefronts of activation radiate into surrounding atrial tissue. Conduction becomes slower and less organized with increasing distance from the rotors, likely because of atrial structural remodeling, resulting in fibrillatory conduction. Interestingly, the dominant rotors in AF appear to cluster primarily in the junction between the LA and PVs. One study also demonstrated that the PV-LA region has heterogeneous EP properties capable of sustaining reentry. As noted earlier, autonomic inputs can be important in triggering and maintaining AF, and many of these inputs are clustered close to the PV-LA junction.


The role of PVs in the initiation and perpetuation of persistent AF seems less prominent than in the setting of paroxysmal AF, likely secondary to the electrical and structural remodeling associated with persistent AF. Non-PV triggers occur more commonly, and the reentry sites required for AF perpetuation are more often found outside the PV-LA junction in persistent AF than in paroxysmal AF.


Pulmonary Vein Anatomy


PVs can have variable anatomy. Most hearts examined are found to have four PVs with distinct ostia. The PV ostia are ellipsoid with a longer superoinferior dimension, and funnel-shaped ostia are frequently noted in patients with AF. The right superior PV lies just behind the SVC or RA, and the right inferior PV projects horizontally. The left superior PV lies between the LA appendage (LAA) and the descending aorta, and the left inferior PV courses near the descending aorta ( Fig. 15.5 ). The superior PVs project forward and upward, whereas the inferior PVs project backward and downward. PVs are larger in patients with AF than in normal subjects, in men than in women, and in persistent versus paroxysmal AF.




Fig. 15.5


Segmented Cardiac Computed Tomography Images Demonstrating Anatomic Relationship of the Left Atrium (LA) and Pulmonary Veins (PVs).

Images are viewed from multiple projections: right lateral (RL) , right anterior oblique (RAO) , anteroposterior (AP) , left anterior oblique (LAO) , left lateral (LL) , and posteroanterior view (PA) . AO, Aorta; IVC, inferior vena cava; LAA, LA appendage, LI, left inferior PV; LPA, left pulmonary artery; LS, left superior PV; LV, left ventricle; PA, pulmonary artery; RA, right atrium; RI, right inferior PV; RPA, right pulmonary artery; RV, right ventricle; RS, right superior PV; SVC, superior vena cava.


Significant variability of PV morphologies can be observed in 40% of patients undergoing AF ablation, including: (1) common ostium (in approximately 25% of patients; Fig. 15.6 ) of left-sided or, less frequently, right-sided PV; (2) supernumerary right PVs (right middle PV is observed in 8% to 29% of patients; see Fig. 15.6 ); (3) anomalous PVs arising from the LA roof; and (4) multiple ramification and early branching (especially of the right inferior PV).




Fig. 15.6


Cardiac Computed Tomography Angiogram (Posteroanterior and Cardioscopic Views) of the Left Atrium and Pulmonary Veins (PVs).

(A and B) The left-sided PVs share a common ostium. (C and D) Three PVs are observed on the right side. LAA, Left atrial appendage; LCPV, left common PV; LSPV, left superior PV; RIPV, right inferior PV; RMPV, right middle PV; RSPV, right superior pulmonary vein.


The PVs are covered by myocardial sleeves formed by one or more layers of myocardial fibers oriented in a circular, longitudinal, oblique, or spiral direction. These sleeves, continuing from the LA into the PV, vary from 2 to 25 mm in length (mean, 13 mm). The length of the myocardial sleeves usually has a distinctive distribution; superior PVs have longer and better developed myocardial sleeves than inferior PVs, which can potentially explain why arrhythmogenic foci are found more often in the superior PVs than in the inferior PVs. It should be noted that all PVs in all individuals have such myocardial sleeves, regardless of the presence or absence of AF.


The walls of the PVs are composed of a thin endothelium, a media of smooth muscle, and a thick outer fibrous adventitia. The transition from atrial to venous walls is gradual because the myocardial sleeves from the LA overlap with the smooth muscle of the venous wall. The myocardial sleeves are thickest at the venoatrial junction (mean, 1.1 mm) and then gradually taper distally. Furthermore, the thickness of the sleeves is not uniform, with the inferior walls of the superior veins and the superior walls of the inferior veins having the thicker sleeves. Throughout the PV, and even at the venoatrial junction, there are gaps in the myocardial sleeves mainly composed of fibrous tissue. The arrangement of the myocyte bundles within the sleeves is rather complex. There appears to be a mesh-like arrangement of muscle fascicles made up of circularly oriented bundles (spiraling around the long axis of the vein) that interconnect with bundles that run in a longitudinal orientation (along the long axis of the vein). Such an arrangement, together with the patchy areas of fibrosis seen, can be relevant to the role of the PVs in the initiation of AF.


Electrophysiology of Pulmonary Vein Musculature


As noted, the PVs play a crucial role in the initiation and, in some cases, maintenance of AF. However, it is not clear what makes this region so susceptible to the arrhythmia. There are, at present, limited data available on the ionic mechanisms that underlie the arrhythmogenicity of PVs. Detailed mapping studies have suggested that reentry within the PVs is most likely responsible for their arrhythmogenicity, although abnormal automaticity and triggered activity have also been observed. However, the rarity with which AF continues within the PV after electrical isolation (see later) calls into question whether the PVs are adequate in themselves to maintain AF.


The EP features of the PV, with its distinct area of slow conduction, decremental conduction, nonuniform anisotropy, and heterogeneous repolarization, create potential substrates for reentry. The heterogeneous fiber orientation in the transition from the LA to the PV sleeve results in unique conduction properties in this area. It is possible that the complex arrangement of muscle fibers within the myocardial sleeves and the uneven distribution of interspersed connective and adipose tissue account for the greater degree of decremental conduction observed in the myocardial sleeves than in the LA and for the heterogeneity in conduction properties and refractory periods among the fascicles in the myocardial sleeves. Therefore the fractionation of PV potentials commonly observed during premature stimulation (which usually indicates local slowing of conduction) is consistent with anisotropic properties that can be attributable to the complex arrangement of muscle fascicles within the myocardial sleeves.


Several studies suggested that abnormal automaticity or triggered activity, either alone or in combination with the reentrant mechanisms described previously, can play a role in the initiation of AF. These studies suggested that the propensity of PVs to exhibit abnormal automaticity or triggered activity is enhanced by pathological conditions. Further work also implicated LA posterior wall in the genesis of AF. Studies suggested that the PVs, together with the posterior LA, have an important role in the persistent form of AF. However, the nature of the relationship between this arrhythmogenic region and the pathological conditions that provide a substrate for AF has not been elucidated. Whether the critical region is the posterior LA, the PVs, or both, has been the source of ongoing debate.


Pulmonary Vein Tachycardia Versus Pulmonary Vein Fibrillation


In patients with paroxysmal AF originating from the PVs, a wide spectrum of atrial arrhythmias can coexist. Extensive monitoring frequently documents coexisting paroxysms of AT and AF. Furthermore, patients with paroxysmal AF usually have multiple foci in multiple PVs, and many of these foci originate distally in those veins.


In patients whose only clinical arrhythmia is PV AT, the clinical course is more comparable to that in patients with AT from other anatomic locations than to patients with PV AF. Those patients demonstrate a largely focal process, without evidence of a more progressive and diffuse disease as observed in the population with paroxysmal AF, and they have no tendency to develop further atrial arrhythmias during long-term follow-up. Notably, when patients with PV AT present with recurrence, in almost all cases this is from the original focus. In contrast, patients with paroxysmal AF have recurrences from foci in other PVs and from within the body of the LA. Importantly, in most patients with PV AT, the focus is located at the ostium of the vein (or within 1 cm of the ostium), rather than from further distally (2 to 4 cm). These observations suggest that patients with focal PV AT can potentially represent a different population from those with PV AF: PV AT patients having a discrete and focally curable process, in contrast to the more diffuse process involving multiple PVs and the LA seen in AF.


The spontaneous onset of focal AT from the PVs and its lack of inducibility with programmed stimulation suggest that this arrhythmia is more likely to be caused by abnormal automaticity or triggered activity rather than reentry. However, attempts to classify the arrhythmia mechanism of focal AT definitively in the EP laboratory are limited because of the significant overlap in the arrhythmia characteristics (initiation, response to drugs).


Genetics in Atrial Fibrillation


It is increasingly recognized that a widespread heritable component underlies AF, especially in patients without apparent risk factors. Studies have shown that at least 5% of all patients with AF and 15% to 20% of those with AF and structurally normal hearts had a positive family history. Family history of AF is associated with a 40% increased risk of first-degree relatives developing AF. The association between family history and risk of AF development is stronger with increased numbers of affected first-degree relatives and younger age.


Several genes and genomic regions linked to AF and its substrate have been identified in families, individuals, and different populations. Genetic factors play a critical role in modulating the risk of the arrhythmia both in rare families with mendelian patterns of AF inheritance and in the general population. Although familial AF may be a monogenetic disorder, nonfamilial AF may be a multigenetic disease in which genetic factors interact with environmental variables.


Classic mendelian genetics and candidate gene approaches have identified AF-causing mutations in genes encoding cardiac ion channels, cardiac gap junctions, signaling molecules, as well as transcription factors. Those channelopathies are commonly associated with other phenotypic manifestations, such the long QT, short QT, or Brugada syndromes, as well as inherited cardiomyopathies. AF is prevalent in cardiac channelopathies and can be the presenting feature in some patients. Studies demonstrated AF prevalence of 2% to 29% in long QT syndrome, 6% to 53% in Brugada syndrome, 18% to 70% in short QT syndrome, and 11% to 37% in patients with catecholaminergic polymorphic ventricular tachycardia (VT). These genetic variants promote AF by either abbreviating atrial refractoriness (facilitating reentry), prolonging atrial action potential duration (facilitating triggered activity), or impairing intercellular coupling (resulting in conduction heterogeneity).


It is noteworthy that a recent study described a rare autosomal recessive atrial cardiomyopathy characterized by progressive fibrosis of the atrial myocardium and clinically manifesting with atrial arrhythmias (including AF), atrial dilation, and potential atrial electrical standstill.


Furthermore, whole-exome sequencing (WES) and genome-wide association studies (GWASs) have identified multiple single-nucleotide polymorphisms (SNPs), a type of common genetic variant, as genetic risks associated with AF in the population. The precise molecular mechanisms underlying these variants remain unclear, owing partly to their presence in noncoding areas of the genome with no known effect on protein expression or function. SNPs presumably act as promoters or enhancers of proximate genes. The closest genes identified with various SNPs are involved in diverse functions, such as in encoding of transcription factors, cytoskeletal and scaffolding proteins, and ion channels.




Epidemiology


AF is the most common sustained arrhythmia encountered in clinical practice, accounting for approximately one-third of hospitalizations for cardiac rhythm disturbances. The worldwide age-adjusted prevalence of AF is estimated at 0.596% in men and 0.373% in women, approximately 33 million people. Each year more than 5 million people develop AF worldwide. The prevalence of AF in the general population in the Western world has been estimated at 0.5% to 2%, although the true prevalence of AF is probably higher given the common occurrence of asymptomatic (subclinical) AF. Furthermore, the frequency of AF is progressively increasing in the general population, likely due to increased longevity and the success in reducing overall cardiovascular mortality, as well as increased prevalence of risk factors for AF (such as hypertension and obesity). In 2010, the prevalence of AF in the United States was estimated to be between 2.7 and 6.1 million. This is expected to rise to about 5.6 to 12 million in 2050. In Europe, AF affects about 8 million people and this number is expected to rise to 18 million by 2060.


AF is a progressive disease. More than half of individuals who experience an initial episode of AF will eventually develop recurrent AF, typically within the first 2 years of follow-up. AF progresses from paroxysmal to persistent (despite antiarrhythmic therapy) in approximately 10% at 1 year, in 25% to 30% at 5 years, and in more than 50% beyond 10 years. Furthermore, progression from paroxysmal and persistent AF to permanent AF occurs in up to 34% of patients at 4 years after the initial diagnosis. Maintenance of NSR becomes progressively more difficult as the duration of persistent AF increases. Only 40% to 60% of patients with persistent AF of less than 1 year’s duration remain in NSR 1 year after initiation of antiarrhythmic drug therapy, despite multiple cardioversions; whereas patients with persistent AF of more than 3 year’s duration have only a 15% likelihood of long-term sinus rhythm. Increased age, diabetes, heart failure, chronic pulmonary disease, and hypertension are among potential predictors of progression to permanent AF. AF occurring in otherwise healthy patients is less likely to progress. Also, AF patients treated with catheter ablation exhibit a substantially lower risk of progression of the arrhythmia.


AF is independently associated with significantly increased morbidity and mortality, including a fivefold increased risk for stroke, a twofold increased risk for dementia, a threefold risk for heart failure, and a twofold increased risk for MI. The mortality rate of patients with AF is approximately double that of patients in NSR and is linked to the severity of underlying heart disease and associated comorbidities. Cardiac causes (sudden cardiac death [SCD], heart failure, and MI) account for almost one-half of deaths in anticoagulated patients with AF, whereas stroke and bleeding only account for approximately 6% of all deaths each as the principal cause. The risk of death appears higher in the presence of heart failure, renal insufficiency, diabetes, advanced age, and male gender.


Atrial Fibrillation Risk Prediction


Several prediction models for new-onset AF have been proposed to help identify high-risk individuals and serve as a benchmark to test potential novel risk factors. The Framingham Heart Study (FHS) and the Atherosclerosis Risk in Communities (ARIC) Study each developed a risk for the prediction of AF. More recently, the Cohorts for Heart and Aging Research in Genomic Epidemiology–Atrial Fibrillation (CHARGE-AF) consortium developed and validated a simple risk model for prediction of incident AF using pooled data (greater than 26,000 individuals) from prospective cohort studies, including the Cardiovascular Health Study, FHS, and ARIC. The variables used in the CHARGE-AF risk prediction score include: age, race, height, weight, systolic blood pressure, diastolic blood pressure, current smoking, use of antihypertensive medication, diabetes, history of MI, and history of heart failure. In addition to these variables, the “augmented” CHARGE-AF prediction score also incorporates the PR interval and ECG-derived LV hypertrophy.


Clinical Risk Factors Predisposing to Atrial Fibrillation


AF can be related to a transient reversible cause, such as thyrotoxicosis, acute MI, acute pericarditis, recent cardiac surgery, acute pulmonary disease, alcohol intake, or electrocution. In these cases, AF generally disappears after treatment of the underlying precipitating condition.


AF is thought to be secondary to underlying structural heart disease in more than 70% of patients and is the final arrhythmic expression of a diverse family of diseases. AF derives from a complex continuum of predisposing factors that appear to involve disease processes that contribute to the triggering of AF (e.g., sympathetic and parasympathetic nervous systems [neurogenic AF], predisposing arrhythmias, or ectopic foci in PVs), increase atrial distention (e.g., valvular heart disease, hypertension, and heart failure), decrease the ratio of atrial myocyte to fibrotic tissue, possibly including an increased rate of apoptotic cell death (e.g., hypertension and ischemic heart disease), disrupt intercellular communications (e.g., pericarditis and edema), increase inflammatory mediators (e.g., pericarditis and myocarditis), or alter energy and redox states that modulate the function of ion channels and gap junctions. Table 15.2 lists cardiovascular and other conditions independently associated with AF.



TABLE 15.2

Cardiovascular and Other Conditions Independently Associated With Atrial Fibrillation














































































































































Characteristic/Comorbidity Association With AF
Genetic predisposition (based on multiple common gene variants associated with AF) HR range 0.4–3.2
Older age HR:
50–59 years 1.00 (reference)
60–69 years 4.98 (95% CI 3.49–7.10)
70–79 years 7.35 (95% CI 5.28–10.2)
80–89 years 9.33 (95% CI 6.68–13.0)
Hypertension (treated) vs. none HR 1.32 (95% CI 1.08–1.60)
Heart failure vs. none HR 1.43 (95% CI 0.85–2.40)
Valvular heart disease vs. none RR 2.42 (95% CI 1.62–3.60)
Myocardial infarction vs. none HR 1.46 (95% CI 1.07–1.98)
Thyroid dysfunction (reference: euthyroid)
Hypothyroidism HR 1.23 (95% CI 0.77–1.97)
Subclinical hyperthyroidism RR 1.31 (95% CI 1.19–1.44)
Overt hyperthyroidism RR 1.42 (95% CI 1.22–1.63)
Obesity HR:
None (BMI <25 kg/m 2 ) 1.00 (reference)
Overweight (BMI 25–30 kg/m 2 ) 1.13 (95% CI 0.87–1.46)
Obese (BMI >31 kg/m 2 ) 1.37 (95% CI 1.05–1.78)
Diabetes mellitus vs. none HR 1.25 (95% CI 0.98–1.60)
Chronic obstructive pulmonary disease RR:
FEV1 ≥80% 1.00 (reference)
FEV1 60%–80% 1.28 (95% CI 0.79–2.06)
FEV1 <60% 2.53 (95% CI 1.45–4.42)
Obstructive sleep apnea vs. none HR 2.18 (95% CI 1.34–3.54)
Chronic kidney disease OR:
None 1.00 (reference)
Stage 1 or 2 2.67 (95% CI 2.04–3.48)
Stage 3 1.68 (95% CI 1.26–2.24)
Stage 4 or 5 3.52 (95% CI 1.73–7.15)
Smoking HR:
Never 1.00 (reference)
Former 1.32 (95% CI 1.10–1.57)
Current 2.05 (95% CI 1.71–2.47)
Alcohol consumption RR:
None 1.00 (reference)
1–6 drinks/wk 1.01 (95% CI 0.94–1.09)
7–14 drinks/wk 1.07 (95% CI 0.98–1.17)
15–21 drinks/wk 1.14 (95% CI 1.01–1.28)
>21 drinks/wk 1.39 (95% CI 1.22–1.58)
Habitual vigorous exercise RR:
Nonexercisers 1.00 (reference)
<1 day/wk 0.90 (95% CI 0.68–1.20)
1–2 days/wk 1.09 (95% CI 0.95–1.26)
3–4 days/wk 1.04 (95% CI 0.91–1.19)
5–7 days/wk 1.20 (95% CI 1.02–1.41)

AF, Atrial fibrillation; BMI, body mass index; CI, confidence interval; FEV1, forced expiratory volume in 1 second; HR, hazard ratio; OR, odds ratio; RR, risk ratio.

From Kirchhof P, Benussi S, Kotecha D, et al. 2016 ESC guidelines for the management of atrial fibrillation developed in collaboration with EACTS. Eur Heart J . 2016;37:2893–2962.


The most frequent causes of acute AF are MI and cardiothoracic surgery. The most common clinical risk factors for chronic AF are hypertension and ischemic heart disease, with the subset of patients having congestive heart failure most likely to experience the arrhythmia. In the developing world, hypertension and rheumatic valvular (usually mitral) and congenital heart diseases are the most commonly related conditions.


The presence of SND also predicts an increased risk for AF. Atrial high-rate episodes (usually indicative of AF or AFL) lasting longer than 5 minutes were noted in 29% of the patients receiving pacemakers for SND but with no prior history of AF.


In younger patients, approximately 30% to 45% of paroxysmal cases and 20% to 25% of persistent cases of AF occur in the absence of any chronic or acute risk factors for the arrhythmia, a condition historically referred to as lone AF. However, given the ambiguous definition in the literature and lack of mechanistic or clinical utility, categorizing AF as “lone” or “idiopathic” is potentially confusing and should not be used to guide therapeutic decisions.


Unmodifiable Risk Factors


Age.


The prevalence of AF increases with advancing age. AF is uncommon in childhood except after cardiac surgery. AF occurs in less than 1% of individuals younger than 60 years, but in approximately 6% of those older than 65 years, and in more than 10% of those older than 80 years. The median age of patients with AF is approximately 75 years. About 75% of patients with AF are 65 years of age or older, and 45% of patients with AF are 75 years of age or older. In adults 55 years of age and older, the lifetime risk of the development of AF is approximately 24% in men and 22% in women.


Gender.


The age-adjusted annual incidence of AF is higher in men compared with women (3.8 vs. 1.6 per 1000 person-years). Similarly, the age-adjusted prevalence of AF is higher in men than in women (10.3% vs. 7.4% in adults aged greater than or equal to 65 years). However, gender-related differences in AF incidence and prevalence appear to be related to other risk factors. One report found that, after adjusting for AF-related risk factors, male gender was no longer an independent risk factor for AF. Of note, the lifetime risk for AF is similar between men and women despite higher AF incidence in men, likely related to the shorter life expectancy in men.


Race.


The age-adjusted risk of developing AF is higher in whites as compared to blacks, Asians, and Hispanics.


Genetics.


More than 5% of all patients with AF and 15% to 20% of those with AF in the absence of known causal conditions had a positive family history. Offspring of parents with AF (especially those with early-onset AF with no conventional risk factors) have a two- to threefold greater risk of developing AF. Several genes and genomic regions linked to AF and its substrate have been identified in families, individuals, and different populations. However, whether genetic profiles can be constructed to identify at-risk populations and whether intervention on modifiable risk factors can reduce the risk of AF in populations with genetic predisposition remain to be investigated.


Modifiable Risk Factors


Hypertension.


Hypertensive heart disease is the most common underlying chronic disorder in patients with AF in developed countries. In fact, hypertension has been found to affect 49% to 90% of the participants in major AF clinical trials. Hypertension per se increases the risk of incident AF by about twofold. Furthermore, the coexistence of hypertension and AF increase the risk of stroke, heart failure, hospitalization, and overall mortality by twofold as compared to AF patients without hypertension. Longstanding hypertension, especially if suboptimally controlled, is associated with sympathetic activation, activation of the renin–angiotensin–aldosterone system, LV hypertrophy, diastolic dysfunction, LA stretch, and structural atrial remodeling, all of which can contribute to development and progression of AF. In addition, hypertension commonly coexists with multiple morbidities that also increase the risk of AF, such as heart failure, coronary artery disease, diabetes, and obesity.


Of note, longitudinal patterns or long-term trajectories of systolic blood pressure, pulse pressure, and hypertension treatment were associated with higher 15-year risk of AF development, and performed better than single time-point measurements, especially where prolonged hypertension is present and antihypertensive treatment is used. Interestingly, distinct trajectories for diastolic blood pressure were not associated with increased 15-year risk of AF.


Heart failure.


Heart failure and AF share several risk factors and common pathophysiologic processes. In a global AF registry, heart failure was prevalent in 33% of paroxysmal, 44% of persistent, and 56% of permanent AF patients. In the Framingham study, 41% of patients with AF and heart failure developed heart failure first, 38% developed AF first, and in the remaining 21% AF and heart failure occurred at the same time. On the other hand, AF develops in up to 42% of heart failure patients. The prevalence of AF increases with heart failure severity, ranging from 5% in functional class I patients compared with approximately 50% in class IV patients. Among a cohort of heart failure patients with preserved left ventricular ejection fraction (LVEF), 29% had a history of AF, and AF or AFL was documented in 65% of those presenting with acute diastolic heart failure.


Patients with concomitant heart failure and AF exhibit a grim prognosis, with a 1-year mortality of 9.5% and worsening of heart failure in almost 25%. In heart failure patients undergoing implantable cardioverter-defibrillator (ICD) implantation, a history of AF at time of procedure identifies additional risk of heart failure and death, and the new detection of AF afterward is associated with even higher rates of death.


In addition to their shared underlying risk factors, AF and heart failure also are independent risk factors for one another. AF can be both a cause and a consequence of heart failure. Heart failure is a potent risk factor for incident AF, with a sixfold increase in the risk of developing AF, while AF is associated with a threefold increased risk of incident heart failure. AF can cause impairment of systolic and diastolic ventricular function not present in sinus rhythm. The loss of atrial systole as well as AF-induced impaired atrial function and atrial fibrosis result in reduced LV filling and cardiac output and can be a direct cause of diastolic heart failure. In addition, irregular and rapid ventricular rates in AF can lead to LV systolic dysfunction (tachycardia-induced cardiomyopathy). AF is also associated with LV myocardial fibrosis, which contributes to diastolic dysfunction. On the other hand, increased ventricular filling pressures and afterload, functional valvular regurgitation, and activation of the renin–angiotensin–aldosterone system in heart failure can lead to atrial stretch, fibrosis, and dilation. In addition, altered calcium handling and calcium overload in heart failure can promote initiation and perpetuation of AF.


Valvular heart disease.


AF has been conventionally classified into “valvular” or “nonvalvular” AF. While the 2012 ESC Guidelines defined valvular AF as rheumatic valvular disease (predominantly mitral stenosis) or prosthetic heart valves, the 2014 AHA/ACC/HRS guidelines defined nonvalvular AF as AF in the absence of rheumatic mitral stenosis, or a mechanical heart valve, but explicitly added bioprosthetic heart valves or mitral valve repair.


A wide spectrum of valvular lesions can lead to the development of AF. The major independent risk factor is LA enlargement, which is more likely to result from mitral valve disease. Elevated LA pressure causes myocardial stretch and atrial remodeling, which promote AF.


Rheumatic heart disease, now uncommon in developed countries, remains an important underlying substrate for AF in the developing world. The prevalence of AF in these patients varies with the type of valve disease, occurring in only 1% of patients with aortic valvular disease, 16% of those with isolated mitral regurgitation, 29% of patients with isolated mitral stenosis, and 52% of those with combined mitral stenosis and regurgitation. In addition, the risk of AF is known to correlate with LA size; the incidence of AF rises from 3% when the LA diameter is less than 40 mm to 54% if the LA diameter exceeds 40 mm. Patients with mitral stenosis and AF are at a particularly high risk for thromboembolic events and have been excluded from any further trials studying anticoagulation regimens for AF. The risk of thromboembolism is higher for AF patients with mitral than those with aortic valve disease.


Nonrheumatic valvular heart disease is common in patients with “nonvalvular” AF (i.e., valve disease with neither rheumatic mitral stenosis nor valve prosthesis), with reported prevalence of up to 25%, especially those with permanent AF. The majority of these patients have mitral regurgitation (61%), aortic regurgitation (24%), and aortic stenosis (32%). Of note, an increased thromboembolic risk has been observed in nonvalvular AF patients who have nonrheumatic native valve disease compared with those without valve disease; however, neither the valve disease per se nor its severity was clearly associated with this risk. Those patients were older and had a higher CHA2DS2-VASc score, which was likely to explain the higher embolic risk.


Coronary artery disease.


Coronary artery disease is present in more than 20% of patients with AF. In contrast, the prevalence of AF in the total population with chronic stable coronary artery disease is relatively low. Nonetheless, AF occurs transiently in 6% to 22% of patients with acute coronary syndromes, and its occurrence in this setting is associated with increased short- and long-term morbidity and mortality. Ischemic heart disease can predispose to the development of AF by atrial stretch and dilation secondary to ischemia-related diastolic and systolic ventricular dysfunction as well as atrial ischemia.


Recent evidence suggests that AF patients are at substantially increased risk (by 47%) of incident MI compared with those without AF. The relative risk is particularly higher in patients free of coronary artery disease at baseline (by 71%), in those younger than 60 years, and in women. Several mechanisms have been proposed to account for the increased risk of MI in AF patients: (1) AF-induced prothrombotic state caused by increased systemic platelet activation, thrombin generation, and endothelial dysfunction; (2) AF-mediated inflammation that can potentially promote plaque rupture and MI; (3) direct coronary thromboembolism from LAA clots; (4) demand ischemia related to fast ventricular rates during AF; and (5) presence of common risk factors that predispose to the development of both AF and coronary artery disease (e.g., diabetes, hypertension, advanced age).


Congenital heart disease.


During long-term follow-up, AF develops in more than one-third of patients with congenital heart disease. AF is more common in patients with severe congenital defects, residual left-sided lesions, or unrepaired heart disease. AF is frequently associated with markers of left-sided heart disease (i.e., LV systolic dysfunction and LA dilation) and is most commonly seen in patients with congenital aortic stenosis, mitral valve disease, palliated single ventricles, unrepaired heart defects, or end-stage heart disease. Compared to patients without congenital heart defects or with simple congenital heart defects, AF develops at a younger age in patients with complex congenital heart defects. AF is rarely seen in patients with atrial septal defects before the age of 40 years, but the incidence can approach 50% in unrepaired patients beyond 60 years of age.


Coexistence of episodes of AF and macroreentrant atrial tachycardia (MRAT) has been reported in a considerable number of patients (33%) with congenital heart disease. Regular ATs preceded development of AF in about two-thirds of patients. Approximately 30% of patients who have previously undergone successful catheter ablation for MRAT develop AF during long-term follow-up.


Obstructive sleep apnea.


Accumulating evidence demonstrates an independent association between AF and obstructive sleep apnea. AF occurs in 5% of individuals with severe sleep apnea and only 1% of those without sleep apnea. The prevalence of at least moderate sleep apnea was reported in up to 32% of patients with AF and structurally normal hearts. Furthermore, cross-sectional studies demonstrated that patients with AF had a significantly higher risk of obstructive sleep apnea than matched controls (49% vs. 33%), and multivariate analysis demonstrated a strong independent association between sleep apnea and AF (odds ratio, 2.2). In addition, several prospective studies showed that obstructive sleep apnea predicts the occurrence of future AF. Untreated obstructive sleep apnea was associated with a remarkably high rate of AF recurrence at 1 year compared with patients with unknown sleep apnea status (83% vs. 53%). The risk of AF in patients with obstructive sleep apnea appears to increase linearly along with the severity of obstructive sleep apnea. In some patients with obstructive sleep apnea and paroxysmal AF, paroxysms of AF were found to be nocturnal and, at least in part, temporally related to respiratory obstructive events.


Obstructive sleep apnea can affect AF response to medical and catheter ablative therapies. Patients with obstructive sleep apnea can have 25% to 31% increased risk for AF recurrence after catheter ablation in comparison with patients with no sleep apnea. On the other hand, the use of continuous positive airway pressure (CPAP) therapy is associated with more than 40% relative risk reduction in AF recurrence in patients with obstructive sleep apnea, regardless of the AF treatment strategy (medical therapy or catheter ablation).


Several mechanisms are proposed by which obstructive sleep apnea increases the risk of AF. Repetitive forced inspiration against a collapsed upper airway during apneic episodes generates substantial shifts in intrathoracic pressure, with consequent increase of venous return, leading to increased RV preload and LV afterload. Pressure and volume overload causes stretch of the atria and PV ostia. Acute atrial stretch can shorten atrial refractoriness (possibly by opening of stretch-activated ion channels) and impair atrial conduction. Repeated atrial stretch can lead to structural atrial remodeling with dilation and fibrosis. In addition, repetitive apneas and hypopneas are accompanied by intermittent hypoxemia and hypercapnia, which cause chemoreceptor-induced sympathetic activation and parasympathetic withdrawal. Sympathetic hyperactivity (which is further enhanced by recurrent arousal) results in peripheral vasoconstriction and systemic hypertension, as well as increased heart rate, and reduced heart rate variability. Also, intermittent hypoxia and postapneic reoxygenation induce oxidative stress and inflammatory processes contributing to LA remodeling and fibrosis. These mechanisms can act as both triggers and perpetuators of AF, and can predispose to arrhythmia recurrence and affect response to different pharmacologic and nonpharmacologic therapies for AF. CPAP therapy is likely to effectively reverse these mechanisms and thereby decrease AF occurrence. Finally, obstructive sleep apnea often coexists with multiple morbidities that also increase the risk of AF, such as heart failure, diabetes, obesity, and systemic hypertension.


Obesity.


Obesity (body mass index [BMI] greater than 30 kg/m 2 ) is associated with a significantly higher risk of AF. Overweight populations have higher incidence, prevalence, severity, and progression of AF compared with their normal weight counterparts. A recent meta-analysis found that for every 5 kg/m 2 increase in BMI, there were 10% to 29% greater risks of incident, postoperative, and postablation AF. In overweight and obese individuals with symptomatic AF, progressive long-term sustained weight loss has a dose-dependent effect on long-term freedom from AF. Notably, weight fluctuation of greater than 5% had an adverse effect on overall freedom from AF, with a twofold greater likelihood of recurrent arrhythmia. A recent meta-analysis found a 3.1% greater risk of recurrent AF postablation for every one unit increase in BMI.


Obesity is often associated with cardiometabolic comorbidities, such as hypertension, diabetes mellitus, autonomic dysfunction, and sleep apnea, all of which can generate a susceptible AF substrate. However, obesity also appears to be a distinct risk factor for AF, independent of the coincidental accumulation of other comorbidities. Obesity was linked to higher risk of AF even among young and seemingly healthy individuals. Several mechanisms have been proposed to explain the association between obesity and AF. Progressive weight gain results in electrical and structural atrial remodeling, atrial interstitial fibrosis, increased LA pressure and volume, ventricular hypertrophy and impaired diastolic function, low-grade systemic inflammation, and myocardial lipidosis. Obesity has also been associated with increased epicardial fat thickness, which has been associated with AF, likely due to altered atrial electrophysiology and autonomic imbalance. On the other hand, weight loss is associated with reversal of atrial dilation and LV hypertrophy.


Diabetes.


Diabetes appears to confer an increased risk for the development of AF. In the FHS, diabetes was associated with 40% higher risk of AF in men and 60% higher risk in women after 38 years of follow-up. Other studies found that AF was more prevalent in subjects with pre-diabetes compared to controls and correlated positively with hemoglobin A1c. Notably, intensive glycemic control does not affect the rate of new-onset AF.


Several mechanisms can potentially underlie the association of diabetes and AF. Diabetes-related cardiomyopathy leads to ventricular systolic or diastolic dysfunction, increased filling pressures, and atrial remodeling. In addition, cardiac autonomic neuropathy has been implicated by leading to sympathetic overactivity and neural remodeling. Furthermore, insulin resistance can be associated with a proinflammatory environment within the myocardium.


Hyperthyroidism.


The prevalence of AF in patients with overt hyperthyroidism ranges between 10% and 15%, and the risk is approximately sixfold that of the euthyroid population. Also, subclinical hyperthyroidism is a risk factor associated with a threefold increase in development of AF. The risk of AF is higher in men, in the elderly, and in patients with triiodothyronine (T3) toxicosis.


About two-thirds of the patients with AF due to hyperthyroidism revert spontaneously to NSR after treatment of the thyrotoxic state, typically within 3 to 6 months of becoming euthyroid. Even though hyperthyroidism is considered a reversible cause of AF, the arrhythmia persists or recurs in up to 30% to 40% of patients after restoring a euthyroid status. Older age (greater than 55 years), long duration of hyperthyroidism (greater than 5 years), long duration of pretreatment AF, severe LV dysfunction, and LA enlargement are independent predictors for continued AF following the successful treatment of hyperthyroidism.


Among all patients with new-onset AF, less than 1% of AF incidence is caused by overt hyperthyroidism. Although the yield of abnormal thyroid function testing in these patients is low, the benefit associated with the ability to restore patients with thyrotoxicosis to a euthyroid state and NSR justifies thyroid-stimulating hormone (TSH) testing in most patients with recent onset of otherwise unexplained AF.


The mechanisms by which hyperthyroidism enhances vulnerability to AF likely include abbreviation of the atrial action potential duration and refractory period, increased atrial ectopic activity, elevation of LA pressure secondary to increased LV mass and impaired ventricular relaxation, and myocardial ischemia resulting from increased resting heart rate.


Pulmonary embolism.


AF can occur in the setting of acute pulmonary embolism, likely secondary to acute RV pressure overload and subsequent RA dilation. AF can be seen as a presenting sign, during the early phase, or later in the course of recovery from pulmonary embolism. Furthermore, patients with prior pulmonary embolism exhibit a substantially increased (ninefold) incidence of late onset AF.


Of note, some studies have suggested that AF can potentially be a cause, rather than a consequence, of pulmonary embolism. The prothrombotic state associated with AF can potentially promote RA thrombus formation with subsequent embolization to the lungs. However, this hypothesis has yet to be confirmed.


Chronic kidney disease.


Chronic kidney disease (CKD) increases the risk of the development of AF. Among patients with chronic kidney disease, the prevalence of AF is two- to threefold higher than reported in the general population. Moreover, AF prevalence increases in a dose-dependent fashion as renal function worsens. In one report, the prevalence of AF was 1.0% among adults without CKD, and 2.8%, 2.7%, and 4.2% among adults with stage 1 to 2, stage 3, and stage 4 t–5 CKD, respectively. Other measures of kidney dysfunction, such as albuminuria, are also associated with higher AF risk. Importantly, incident AF is independently associated with increased risk of progression to end-stage renal disease requiring dialysis in adults with chronic kidney disease (likely due to the proinflammatory, profibrotic, and prothrombotic state as well as altered hemodynamics associated with AF).


Several possible mechanisms can potentially explain the high rate of identified AF among patients with chronic kidney disease, including a high prevalence of shared risk factors between chronic kidney disease and AF (e.g., advanced age, LV hypertrophy, hypertension, diabetes), increased systemic inflammation, sympathetic activation, myocardial fibrosis, and activation of the renin–angiotensin–aldosterone system.


Exercise and fitness.


Recent data describing the intensity, duration, and frequency of exercise suggest a U -shaped relationship with AF and mortality. Sedentary lifestyle significantly increases the risk of AF, while moderate-intensity exercise is protective against future AF in both men and women. However, vigorous long-term exercise training has a gender-specific association with AF risk. Compared with sedentary counterparts, men who exercised at a vigorous intensity had a threefold risk of incident AF. In contrast, women exercising at vigorous intensities actually had a lower incidence of AF.


The mechanisms of AF promotion by endurance exercise remain speculative. Proposed contributors include atrial stretch and dilatation, LV hypertrophy, chronic systemic inflammation, increased vagal tone, sinus node remodeling, anatomic adaptation, and illicit drugs. However, it remains unclear why intense physical activity increases the AF risk only in men and not in women. Of note, AF in athletes is less likely to be associated with the common risk factors for AF identified in the general population. On the other hand, physical inactivity predisposes to AF risk factors (e.g., hypertension, obesity, and diabetes), and is associated with systemic inflammation and sympathetic activation.


Alcohol.


AF is the most common arrhythmia associated with alcohol consumption. Importantly, the total amount and pattern of alcohol drinking as well as the type of alcoholic beverage appear to impact AF risk. Habitual moderate (7 to 21 standard drinks/wk) and heavy (greater than 21 standard drinks/wk) alcohol consumption, even after correcting for binge drinking, increases the incidence of AF in a dose-dependent manner, with an 8% increase in AF risk for each 1 standard drink per day (or 12 g pure alcohol per day) increase of alcohol consumption.


In addition, an association exists between binge-pattern drinking (greater than 5 drinks on a single occasion) and increased AF risk, independent of the number of drinks consumed per week. AF in the setting of acute consumption is well recognized, occurring in up to 60% of binge drinkers. In fact, the “holiday heart syndrome” describes AF occurring following weekends or holidays when alcohol intake is increased. Furthermore, the risk for AF was found to be most pronounced with liquor, modest for wine, and no excess risk was detected with beer.


Among patients with a history of AF, alcohol consumption is associated with an increased risk of progression from paroxysmal to persistent AF as well as increased risk of AF recurrence following catheter ablation. A safe level of daily alcohol consumption in AF patients has not been identified.


Several mechanisms have been implicated in mediating the adverse effects of alcohol. Ethanol can lead to electrical atrial remodeling, resulting in slowing of intraatrial conduction and abbreviation of atrial refractoriness. Other potential mechanisms include sympathetic stimulation, modulation of vagal tone, alterations in oxidative stress, electrolyte imbalances (hypokalemia, hypomagnesemia), and alcohol-induced cardiomyopathy. Furthermore, ethanol and its metabolite, acetaldehyde, have direct cardiotoxic effects, including direct effects on atrial excitation-contraction coupling, inhibition of calcium release from the sarcoplasmic reticulum, generation of oxidative stress, accelerated protein catabolism, and derangements in fatty acid metabolism and transport.


Smoking.


Several, but not all, studies demonstrated an association between past and current cigarette smoking and an increased risk of AF. In one report, current smokers exhibited almost twice the risk of AF as compared to never smokers. Past smokers also had an increased risk of AF, albeit lower than those who continued to smoke. Furthermore, the cumulative amount of smoking in cigarette-years was correlated with an increased risk of developing AF. In addition, second-hand smoking, particularly when present during development and early childhood, was statistically significantly associated with the presence of AF. This relationship was particularly strong in the absence of known AF risk factors. Of note, smoking has also been reported to predict worse outcome (i.e., increased risk of intracranial bleeding, mortality, and the combined outcome of stroke or death) in patients with AF.


Several mechanisms can be involved in the associations of smoking with AF. Nicotine itself has been linked to cardiac arrhythmias, including AF, likely secondary to sympathetic activation, atrial electrical alterations, atrial fibrosis, and atrial structural remodeling. Furthermore, carbon monoxide can influence cardiac automaticity. In addition, oxidant substances and polycyclic aromatic hydrocarbons can potentially play a role.


Caffeine.


Caffeine, a methylxanthine compound that is chemically similar to theophylline, increases neurohormonal and sympathetic stimulation. Therefore caffeine has been addressed as a potential trigger for AF. However, studies failed to demonstrate any significant relationship between habitual or heavy caffeine consumption and incident AF.


Recreational drugs.


Data on recreational (illicit) drugs as risk factors for AF per se are sparse. AF has not been reported to be associated with amphetamine, heroin, or LSD abuse. Limited reports suggest a potential effect of the abuse of cannabis, cocaine, ecstasy, and anabolic androgenic steroids on AF.


Cannabis, the most commonly used recreational drug, has been associated with several cases of AF (starting within minutes to 3 hours of cannabis use) in young people without comorbidities. The underlying mechanism probably is related to sympathetic activation and reduced coronary microcirculation.


Drug-Induced Atrial Fibrillation


Several cardiovascular and noncardiovascular drugs can induce AF. The overall incidence of drug-induced AF is relatively low; however, since drug-induced AF in most cases is paroxysmal, spontaneously terminating in a few minutes or hours, the true incidence of drug-induced AF is probably underestimated.


Transient AF is observed in approximately 3% of patients receiving adenosine for treating SVT and up to 17% of those receiving adenosine during EP studies. Also, AF can be induced by positive inotropic agents such as dobutamine for stress echocardiography (0.4% to 2%) and milrinone (5%). Furthermore, the use of dopamine, or dobutamine following cardiac surgery is associated with a higher incidence of postoperative AF. Paroxysmal AF is a relatively common complication (17%) following intracoronary injection of acetylcholine for the provocation of coronary spastic angina.


Several antineoplastic agents can induce AF, including intrapericardial cisplatin (12% to 32%), cyclophosphamide (2%), anthracyclines (1% to 10%), IL-2 (4% to 8%), melphalan (6% to 12%), 5-fluorouracil (1%), and paclitaxel (1% to 1.7%). Other drugs reportedly implicated in precipitating AF include nonsteroidal antiinflammatory agents, high-dose corticosteroids, ondansetron (an antiemetic agent), aminophylline, theophylline, antipsychotic agents (e.g., clozapine, olanzapine), antidepressants (e.g., fluoxetine, trazodone), bisphosphonates (e.g., alendronate), and ivabradine.


Several classes of drugs can induce AF through very different mechanisms, including: (1) alteration of atrial EP properties, with increased focal activity, shortened action potential duration and refractoriness, or reduced conduction velocity (e.g., adenosine, parasympathomimetics, sympathomimetics, and theophylline); (2) adrenergic or vagal stimulation (e.g., acetylcholine, adenosine, sympathomimetics); (3) direct cardiotoxicity causing myocardial fibrosis, cardiomyopathy, myocarditis, or pericarditis (e.g., cancer chemotherapy); (4) myocardial ischemia secondary to coronary vasospasm, thrombosis, or arteritis (e.g., acetylcholine, chemotherapy agents, ondansetron, and sumatriptan); (5) electrolyte disturbances (e.g., diuretics, glucocorticoids); (6) abnormalities in calcium handling (e.g., inotropic agents); (7) release of proinflammatory cytokines (e.g., IL-2); and (8) increased oxidative stress (e.g., cancer chemotherapy).


Postoperative Atrial Fibrillation


Epidemiology


Postoperative AF complicates cardiac surgery in 15% to 63% of patients. The risk of AF is highest among patients undergoing combined coronary artery bypass grafting and mitral valve replacement (63%) and the lowest among patients undergoing isolated coronary revascularization surgery (15% to 40%) and cardiac transplantation (11% to 24%). Postoperative AF has also been known to complicate noncardiac surgery (incidence, 0.3% to 13.7%), especially thoracic and large colorectal surgeries.


Clinical risk factors for the development of postoperative AF include advanced age (greater than 65 years), male gender, Caucasian race, hypertension, prior AF, mitral valve disease, heart failure, LV hypertrophy, diastolic dysfunction, increased LA size, history of MI, withdrawal of beta-blocker therapy, obesity, low BMI, chronic obstructive pulmonary disease, anemia, prolonged PR interval, diabetes, renal dysfunction, tobacco use, and high baseline CRP levels. The CHADS2 and CHA2DS2-VASc scores were also found to be predictive of AF after cardiac surgery.


Several factors related to the surgical procedure also potentially contribute to the development of AF. These include operative trauma from surgical dissection and manipulation, pericardial lesions, atrial dilation (caused by LV dysfunction and intraoperative volume overload), perioperative use of catecholamines, parasympathetic activation, and electrolyte imbalances.


The onset of postoperative AF peaks at 24 to 72 hours after surgery, and declines to 2% at discharge. Among patients with no prior history of AF, postoperative is usually self-limited, terminating within 24 hours in the majority of patients, with a mean duration of about 11 to 12 hours. However, despite its usually transient nature, the arrhythmia is not limited to the early postoperative phase, and can recur between day 6 and day 30 after the operation in about 25% of patients.


Prognosis


The occurrence of postoperative AF is associated with a twofold increase in cardiovascular mortality and morbidity. Postoperative AF is associated with a two- to fourfold increase in stroke risk at 30 days, and is an important predictor of in-hospital and long-term mortality. Numerous postoperative complications have been correlated with postoperative AF, including congestive heart failure, stroke, bleeding complications (from anticoagulation), renal insufficiency, infection, ventricular arrhythmias, prolonged ventilation, reintubation, readmission to the intensive care unit, and prolongation of hospital stay (by about 4 to 5 days). However, it is important to note that a causal relationship between postoperative AF and its associated adverse outcomes is still not well defined.


Mechanism


Although the exact pathophysiology of postoperative AF remains incompletely understood, both acute perioperative and chronic factors appear to play an important role in the initiation and maintenance of the arrhythmia. Acute perioperative factors that have been implicated in the creation of atrial susceptibility to AF include pericardial inflammation, heightened sympathetic tone, use of inotropes, acute atrial injury, ischemia, and oxidative stress, as well as acute atrial dilation secondary to pressure or volume overload. Also, it is likely that a preexisting atrial electrical and structural arrhythmogenic substrate increases the vulnerability to the development of AF when subjected to the acute perioperative stress. In fact, the majority of patients with postoperative AF have underlying atrial disease. Further, the presence of preexistent atrial substrate likely explains the increased risk for developing future AF among patients with new-onset postoperative AF, suggesting that the development of postoperative AF can be a surrogate for an underlying cardiac substrate.




Clinical Presentation


Symptomatic Atrial Fibrillation


AF can be symptomatic or asymptomatic, even in the same patient. Symptoms associated with AF vary, depending on the ventricular rate, the underlying functional status, the duration of AF, the presence and severity of structural heart disease, and the individual patient’s perception.


The hemodynamic consequences of AF are related to loss of coordinated atrial contraction, rapid ventricular rate, and irregularity of ventricular rhythm (independent of rate), as well as long-term consequences such as atrial and ventricular cardiomyopathy. Loss of effective atrial contraction can potentially reduce cardiac output by 15% to 25%. These consequences are magnified in the presence of impaired diastolic ventricular filling, hypertension, mitral stenosis, LV hypertrophy, and restrictive cardiomyopathy. Irregularity of the cardiac cycle, especially when accompanied by short coupling intervals, and rapid heart rates in AF can lead to reduction in diastolic filling, stroke volume, and cardiac output.


Most patients with AF complain of palpitations, chest discomfort, dyspnea, generalized fatigue, or dizziness, although significant interindividual and intraindividual variability exists. Although palpitation, or awareness of the irregularity of the heartbeat, is prominent in more than half of patients with AF (more common in those with paroxysmal AF), its correlation with documented arrhythmia is unimpressive. Dyspnea and fatigue can result in significant activity intolerance.


Chest pain can be related to demand ischemia secondary to reduced cardiac output during AF in patients with coronary artery disease; however, chest pain can occur in AF patients despite the absence of coronary artery disease, potentially related to impaired microvascular flow. Furthermore, AF with a chronically rapid heart rate (more than 120 to 130 beats/min) can lead to tachycardia-mediated cardiomyopathy and heart failure.


Syncope is an uncommon complication of AF that can occur on termination of the arrhythmia in patients with SND or (especially at the onset of an episode) because of rapid ventricular rates in patients with hypertrophic cardiomyopathy, aortic stenosis, or ventricular preexcitation over a BT. The first presentation of asymptomatic AF can be catastrophic—an embolic complication or acute decompensation of heart failure.


AF is associated with two- to three-times higher risk of cognitive decline and all forms of dementia, including Alzheimer’s disease, senile dementia, and vascular dementia. Potential mechanisms of dementia in AF patients include embolic or hemorrhagic strokes, altered cerebral blood flow in AF, and cerebral micro-bleeds from anticoagulation, oxidative stress, and proinflammatory or prothrombotic status.


In some patients, paroxysmal AF can be classified as either vagal or adrenergic , depending on the types of triggers and the temporal distribution of the arrhythmic episodes. Vagal AF typically occurs in young male patients without structural heart disease and characteristically develops during sleep or postprandial. In contrast, patients with adrenergic AF are usually older, often with evidence of underlying heart disease, and AF episodes usually occur during the day and are associated with physical or emotional stress. In patients with paroxysmal AF, the prevalence of vagal AF probably ranges between 6% and 25%, whereas that of adrenergic AF ranges between 7% and 16%. Pure adrenergic and vagotonic forms of paroxysmal AF are uncommon. Approximately 12% of patients with paroxysmal AF exhibit features of mixed vagal and adrenergic patterns.


It is important to note that many patients with AF do not complain of palpitations and present primarily with occult cardiac symptoms, such as fatigue and effort intolerance. Such complaints should not be dismissed as “unrelated,” and those patients should not be labeled “asymptomatic.” On the other hand, many patients with persistent or permanent AF have one or more comorbid conditions (such as sleep apnea, heart failure, pulmonary disease) that can considerably contribute to specific complaints and to overall quality of life. Therefore it is imperative to establish a correlation between any symptoms and AF, as well as ventricular response rates. The effect of regulation of ventricular rate during persistent AF or conversion to NSR on a patient’s symptoms and quality of life can help assess the relative contribution of AF to the patient’s complaints. This is particularly important when the impact of AF on patient’s symptoms and quality of life is being considered as the indication for therapeutic interventions (such as ablation).


Atrial Fibrillation Symptom Scales


The Canadian Cardiovascular Society Severity in Atrial Fibrillation (CCS-SAF) scale ( Table 15.3 ) and the modified European Heart Rhythm Association (EHRA) symptom scale ( Table 15.4 ) have been developed to describe symptom severity and assess the functional consequences of symptoms in AF patients (analogous to the New York Heart Association [NYHA] congestive heart failure functional class and the CCS angina severity class). These scales can provide objective assessment of the patient’s subjective state, help guide symptom-orientated treatment decisions, and facilitate longitudinal patient profiling.



TABLE 15.3

Canadian Cardiovascular Society Severity of Atrial Fibrillation Scale
































Step 1: Symptoms



  • Identify the presence of the following symptoms:



  • Palpitation



  • Dyspnea



  • Dizziness, presyncope, or syncope



  • Chest pain



  • Weakness or fatigue

Step 2: Association
Is AF, when present, associated with the foregoing symptoms? For example: Ascertain whether any of the foregoing symptoms are present during AF and are likely caused by AF (as opposed to some other cause).
Step 3: Functionality
Determine whether the symptoms associated with AF (or the treatment of AF) affect the patient’s functionality (subjective QOL).
CCS-SAF Class Definitions
Class 0 Asymptomatic with respect to AF
Class 1 Symptoms attributable to AF have minimal effect on patient’s general QOL



  • Minimal and/or infrequent symptoms, or



  • Single episode of AF without syncope or heart failure

Class 2 Symptoms attributable to AF have minor effect on patient’s general QOL



  • Mild awareness of symptoms in patients with persistent or permanent AF, or



  • Rare episodes (e.g., less than a few per year) in patients with paroxysmal or intermittent AF

Class 3 Symptoms attributable to AF have a moderate effect on patient’s general QOL



  • Moderate awareness of symptoms on most days in patient with persistent or permanent AF, or



  • More common episodes (e.g., more than every few months) or more severe symptoms, or both, in patients with paroxysmal or intermittent AF

Class 4 Symptoms attributable to AF have a severe effect on patient’s general QOL



  • Very unpleasant symptoms in patients with persistent or paroxysmal AF and/or



  • Frequent and highly symptomatic episodes in patients with paroxysmal or intermittent AF, and/or



  • Syncope thought to result from AF, and/or



  • Congestive heart failure secondary to AF


AF, Atrial fibrillation; CCS, Canadian Cardiovascular Society; QOL, quality of life; SAF, severity of atrial fibrillation.

From Dorian P, Guerra PG, Kerr CR, et al. Validation of a new simple scale to measure symptoms in atrial fibrillation: the Canadian Cardiovascular Society Severity in Atrial Fibrillation (CCS-SAF) scale. Circ Arrhythm Electrophysiol . 2009;2:268–275.


TABLE 15.4

The Modified European Heart Rhythm Association Symptom Scale




























Modified EHRA Score Symptoms Description
1 None AF does not cause any symptoms
2a Mild Normal daily activity not affected by symptoms related to AF
2b Moderate Normal daily activity not affected by symptoms related to AF, but patient troubled by symptoms a
3 Severe Normal daily activity affected by symptoms related to AF
4 Disabling Normal daily activity discontinued

AF, Atrial fibrillation; EHRA, European Heart Rhythm Association.

From Kirchhof P, Benussi S, Kotecha D, et al. 2016 ESC guidelines for the management of atrial fibrillation developed in collaboration with EACTS. Eur Heart J . 2016;37:2893–2962.

a EHRA class 2a and 2b can be differentiated by evaluating whether patients are functionally affected by their AF symptoms. AF-related symptoms are most commonly fatigue/tiredness and exertional shortness of breath, or less frequently palpitations and chest pain.



Silent Atrial Fibrillation


Asymptomatic, or silent, AF occurs frequently; approximately one-third of patients with AF and up to 65% of AF episodes have been shown to be asymptomatic. Furthermore, a poor correlation between symptoms and AF has been demonstrated, and perception of AF patients of their prevailing rhythm is often inaccurate. Continuous monitoring with a pacemaker with dedicated functions for AF detection showed that as many as 40% of patients experienced AF-like symptoms in the absence of AF, whereas 38% of patients with a history of AF had episodes of AF lasting more than 48 hours noted at the time of interrogation even though these patients were asymptomatic. Therefore the lack of symptoms should not be equated with the absence of AF, even in patients previously presented with symptomatic episodes of AF.


In individuals with no prior history of AF, silent AF can be noticed incidentally on routine physical examination or preoperative assessment, during active electrocardiographic screening in at-risk populations (e.g., patients with ischemic strokes), or in patients with cardiac implantable electronic devices (pacemakers, defibrillators, loop recorders). Furthermore, AF can be detected by new technologies, such as smartphone cases with ECG electrodes, smart watches, and blood pressure machines with AF detection algorithms. Occasionally, AF is discovered only after a complication attributable to AF (e.g., stroke or congestive heart failure). Up to 30% of patients presenting with cryptogenic strokes are found to have AF that was not previously recognized.


Asymptomatic AF, especially when paroxysmal, is often missed. It is estimated that 10% to 27% of all patients with AF remain undiagnosed due to the lack of symptoms. In the United States, the prevalence of undiagnosed AF is about 1% to 2% in the general population. Importantly, clinically silent AF is similar to symptomatic AF in terms of overall risks of death, cardiovascular death, or thromboembolic events.


Device-Detected Atrial Fibrillation


Implanted pacemakers or defibrillators with atrial leads are capable of continuously monitoring the heart rhythm and detection of high atrial rates, providing a unique opportunity to identify the occurrence and burden of AF and correlating those episodes to patient’s symptoms ( eFig. 15.4 ). The incidence of device-detected atrial high rate episodes varies depending on patient population, programmed device parameters (atrial rate and duration detection thresholds), and length of follow-up duration. In patients without prior history of AF, silent AF (as indicated by device-detected atrial high-rate episodes) is observed in nearly 30% of dual-chamber pacemaker recipients and 25% of cardiac resynchronization device recipients.





eFig. 15.4


Cardiac Device-Detected Atrial High Rate Episodes.

Pacemaker interrogation strips from three different patients with dual-chamber pacemakers. (A) AF is present on the atrial (A) channel and correctly annotated on the marker (M) channel, with an atrial cycle length ranging between 140 and 190 milliseconds. Rapid and irregular ventricular response is observed on the ventricular (V) channel. (B) AF is present on the atrial channel, and the ventricular channel shows a well-controlled, irregular rhythm. On the marker channel, many of the low-amplitude atrial signals are under-sensed, leading to failure to detect atrial arrhythmias. (C) Sinus rhythm is present, but intermittent noise (high-frequency signals observed in the initial two-thirds of the recording) on the atrial channel is erroneously annotated on the maker channel as atrial activity, leading to inappropriate detection of atrial arrhythmias. AP, Atrial paced; AR, atrial-sensed event in the refractory period; AS, atrial sensed; FD, fibrillation detection; VP, ventricular paced; VS, ventricular sensed.


Accuracy of Device-Detected Atrial Fibrillation


Atrial high-rate episodes have been used as a surrogate for AF, and several studies have documented positive correlation between device-identified atrial high-rate episodes and ECG-documented episodes of AF or AFL with a high degree of sensitivity and specificity. However, different trials used different definitions of an atrial high-rate event (i.e., different programmed detection parameters of atrial rate and duration), and a clear consensus definition is still lacking. Nonetheless, employing an atrial rate cut-off of at least 220 beats/min (or greater than 250 beats/min for better specificity) sustained for durations exceeding 5 minutes provides good sensitivity and specificity for clinically confirmed AF (approaching 98% sensitivity and 100% specificity). Shorter cutoffs can lead to over-detection, commonly due to far-field R- and T-wave oversensing.


It is important to interpret device-stored data with caution. False-positive AF detection can result from oversensing (e.g., far-field R- and T-wave oversensing by the atrial lead, electrical interference, myopotentials, or repetitive nonreentrant ventriculoatrial synchrony) (see eFig. 15.4 ). On the other hand, under-detection of atrial activity is not uncommon due to the small amplitude of atrial electrograms during AF. In addition, atrial high-rate episodes are not specific for AF and can be triggered by AT and AFL. Inspection of device-stored electrograms (and not only marker channels) is important to verify the accuracy of the device diagnostics. However, because of the limited memory allocated for intracardiac electrograms, pacing devices may store only limited electrogram data to corroborate that the events labeled as atrial high-rate episodes or automatic mode switch events are indeed AF or AFL.


False-positive AF detection is also frequently encountered with implantable loop recorders with AF detection algorithms. False-positive detections are often caused by noise oversensing, frequent atrial or ventricular premature beats, T-wave oversensing, or sinus arrhythmia. In one report, the accuracy of implantable loop recorders to detect true AF on a per patient basis was 96%; however, when the same analysis was performed on a per episode basis, then the overall accuracy of AF detection was only 48%.


Clinical Implications of Device-Detected Atrial Fibrillation


Several studies have clearly demonstrated that device-detected silent AFs are associated with increased risk of ischemic stroke and systemic embolism. The risk of thromboembolism appears to be related to the duration of AF episodes (or atrial high-rate episodes) as well as the patients’ risk factor profile (CHA2DS2-VASc score).


The critical burden of atrial high-rate episodes beyond which thromboembolic risk is increased and that warrants therapeutic intervention remains to be defined. Several studies attempted to assess the burden of AF that is associated with adverse clinical outcomes, and AF burden thresholds ranging from 5 minutes to 24 hours were found in different reports to have clinical relevance. However, data are sparse on whether treatment with anticoagulation for subclinical device-detected atrial high-rate episodes reduces the risk of stroke to a similar degree as it does in clinical AF. In addition, current clinical practice guidelines for the treatment of device-detected atrial high-rate episodes are lacking. Based on current evidence, it seems appropriate to consider long-term oral anticoagulation for stroke prevention in high-risk patients with atrial high-rate episodes lasting longer than 5 to 6 minutes, when false-positive AF detections are excluded.


Of note, while device-detected silent AF is known to increase stroke risk, there does not seem to be a proximate temporal relationship between device-detected atrial high-rate episodes and the occurrence of strokes. In fact, in the majority of patients of a study population, no AF was detected on device recordings in the 30 days preceding the thromboembolic events. Although these data imply that the mechanism of stroke may not be related solely to the AF episodes, the association between the observation of atrial high-rate episodes and increased risk of stroke has been consistent, and silent AF is viewed as the culprit in cryptogenic stroke in these patients. However, the value of continuous device-detected atrial arrhythmia information to guide therapeutic intervention for stroke prevention remains to be determined. A recent study suggested that a strategy of urgent initiation of anticoagulation based on device-detected AF did not improve outcomes, likely because of temporal dissociation between AF and stroke. Furthermore, withdrawal of anticoagulation in AF patients after arrhythmia-free periods on device interrogation was associated with worse outcome, implying that the decision on long-term anticoagulation in patients with AF should be based on more comprehensive, individualized assessment of risk and benefit rather than temporal incidence of arrhythmias detected by cardiac devices.




Risk of Thromboembolism


AF is a major risk factor for thromboembolism, causing approximately 15% of the ischemic strokes in the United States, 36% of strokes in patients older than 80 years, and up to 20% of cryptogenic strokes. Moreover, cardioembolic strokes caused by AF are large and multiple, often involve bilateral infarcts, and are associated with the highest rates of mortality and permanent disability. Specifically, patients with AF-related stroke show a 50% likelihood of death within 1 year, compared with 27% for strokes not related to AF.


In the FHS, patients with rheumatic heart disease and AF had a 17-fold increased risk of stroke compared with age-matched controls. For nonvalvular AF, the risk of stroke is estimated to be two to seven times that of subjects without arrhythmia, thus resulting in an average incidence of stroke of 5% per year. This rate may increase to 7% per year when silent cerebral ischemic events and transient ischemic attacks are taken into account.


Although patients with AF in the setting of rheumatic valvular disease are expected to be at high risk of stroke, the stroke risk in nonvalvular AF is not homogeneous across the various subgroups of patients. The risk ranges from less than 1.5% per year in otherwise healthy AF patients who are less than 59 years old to more than 10% per year in older patients, especially when AF is associated with specific conditions or comorbidities. Prior history of stroke, transient ischemic attack, or thromboembolism, age, gender, ethnicity, hypertension, diabetes, coronary artery disease, peripheral artery disease, cardiomyopathy, and heart failure are important risk factors.


Previous systematic reviews have not identified AF pattern (paroxysmal, persistent, or permanent) as an important prognostic risk factor for thromboembolism. In fact, AF stroke risk prediction models have in general not included AF type, and current clinical guidelines recommend that decisions regarding oral anticoagulation be made independently of AF pattern. However, recent data suggest that persistent and permanent AF is associated with an almost twofold higher rate of stroke or systemic embolism than paroxysmal AF after adjustment for other independent predictors.


In addition, a range of biomarkers have been identified as potential predictors of thromboembolic events in AF patients. These include markers of thrombosis (von Willebrand factor, D-dimer), myocardial necrosis (troponin), renal function (creatinine clearance, proteinuria), and the natriuretic peptides (N-terminal pro–B-type natriuretic peptide [NT-proBNP], BNP).


Severe findings on transesophageal echocardiography (TEE) have been identified as independent predictors of stroke and thromboembolism, including the presence of an LA thrombus (relative risk, 2.5), complex aortic plaques (relative risk, 2.1), spontaneous echo contrast (relative risk, 3.7), and low LAA velocities (up to 20 cm/s; relative risk, 1.7). Limited data suggest that large LAA dimensions on CMR may predict a higher risk of thromboembolism.


Stroke Risk Stratification


Several prominent risk stratification schemes have been developed to help distinguish patients with AF who are at high risk of ischemic stroke and other systemic thromboembolism from those with a risk sufficiently low that anticoagulation may not be beneficial when considering the associated bleeding risks.


The CHADS2 index, named for a combination of clinical risk factors ( Table 15.5 ), was the first risk stratification scheme to gain widespread acceptance due to its relative ease of use. The CHADS2 system stratifies patients into low- (CHADS2 score of 0), moderate- (score of 1 to 2), and high-risk (score of 3 to 6) categories. The stroke rate per 100 patient-years without antithrombotic therapy increases by a factor of approximately 1.5 for each one-point increase in the CHADS2 score: from 1.9% for a score of 0% to 18.2% for a score of 6. A major limitation of the CHADS2 scheme is the inadequate discrimination of risk. In fact, a large proportion (more than 60%) of patients are classified as having intermediate risk. Furthermore, this risk scheme is not adequately sensitive in identifying AF patients who are truly at low risk; many patients with AF categorized as low-risk by the CHADS2 score still have stroke rates exceeding 1% per year.



TABLE 15.5

CHADS2 Scoring System for Predicting Stroke and Thromboembolism in Atrial Fibrillation































Letter Clinical Characteristic Points
C Congestive heart failure 1
H Hypertension 1
A Age ≥75 years 1
D Diabetes mellitus 1
S2 Stroke, transient ischemic attack, or thromboembolism 2
Maximum points 6


Some of the limitations of the CHADS2 scheme have been addressed by the newer CHA2DS2-VASc scoring system, which incorporates all components of the CHADS2 system but with greater emphasis on age and includes two additional factors: female sex and vascular disease ( Table 15.6 ). The CHA2DS2-VASc score has the major advantage of discriminating risk probability in lower risk patients and has been shown in multiple cohorts to be the best for identifying truly low-risk patients, even in those with a CHADS2 score of 0 ( Table 15.7 ). A low-risk score is defined as a CHA2DS2-VASc score of 0, an intermediate risk is defined as a score of 1, whereas a high-risk CHA2DS2-VASc score is defined by a score of 2 or greater.



TABLE 15.6

CHA2DS2-VASc Scoring System for Predicting Stroke and Thromboembolism in Atrial Fibrillation











































Letter Clinical Characteristic Points
C Congestive heart failure or left ventricular dysfunction 1
H Hypertension 1
A2 Age ≥75 years 2
D Diabetes mellitus 1
S2 Stroke, transient ischemic attack, or thromboembolism 2
V Vascular disease (prior myocardial infarction, peripheral artery disease, or aortic plaque) 1
A Age 65–74 years 1
S Sex category (i.e., female gender) 1
Maximum points 9


TABLE 15.7

Stroke or Other Thromboembolism Events Based on the CHA2DS2-VASc Scoring System









































































CHA2DS2-VASc Score COHORT OF PATIENTS ON ANTICOAGULATION COHORT OF PATIENTS OFF ANTICOAGULATION
Patients (n = 7239) Adjusted Stroke Rate a
(%/year)
Patients (n = 1084) Adjusted Stroke Rate b
(%/year)
0 1 0 103 0
1 422 1.3 162 0.7
2 1230 2.2 184 1.9
3 1730 3.2 203 4.7
4 1718 4.0 208 2.3
5 1159 6.7 95 3.9
6 679 9.8 57 4.5
7 294 9.6 25 10.1
8 82 6.7 9 14.2
9 14 15.2 1 100

CHA2DS2-VASc, updated version of the CHADS2 ( c ongestive heart failure, h ypertension, a ge, d iabetes, and s troke [doubled]) system, with additional risk factors.

Data from Lip GY, Nieuwlaat R, Pisters R, et al. Refining clinical risk stratification for predicting stroke and thromboembolism in atrial fibrillation using a novel risk factor-based approach. Chest . 2010;137:263–272.

a Theoretical thromboembolism rates without anticoagulation therapy: assuming that warfarin provides a 64% reduction in thromboembolic risk. Data from Lip GY, Frison L, Halperin JL, Lane DA. Identifying patients at risk of stroke despite anticoagulation. Stroke. 2010;41:2731–2738.


b Theoretical thromboembolism rates without antiplatelet therapy: assuming that aspirin provides a 22% reduction in thromboembolic risk.



The R2CHADS2 risk model has emerged from an analysis of the ROCKET-AF (Rivaroxaban Once Daily Oral Direct Factor Xa Inhibition Compared with Vitamin K Antagonism for Prevention of Stroke and Embolism Trial in Atrial Fibrillation) population and was validated in an ATRIA (Anticoagulation and Risk Factors in Atrial Fibrillation) population. In addition to incorporating the same components of the CHADS2 score, the R2CHADS2 scheme awards 2 points for renal dysfunction.


The ATRIA score contains elements of R2CHADS2 but importantly gives different scores for age ranges that vary according to whether the patient had also suffered a stroke or transient ischemic attack ( Table 15.8 ). In one report, the ATRIA score outperformed CHADS2 and CHA2DS2-VASc risk scores largely because its use resulted in an appropriate downward classification (toward no risk).



TABLE 15.8

Anticoagulation and Risk Factors in Atrial Fibrillation Stroke Risk Score




















































Risk Factor Points Without Prior Stroke Points With Prior Stroke
Age, years
≥85 6 9
75–84 5 7
65–74 3 7
<65 0 8
Female 1 1
Diabetes mellitus 1 1
CHF 1 1
Hypertension 1 1
Proteinuria 1 1
eGFR <45 or ESRD 1 1

CHF, Congestive heart failure; eGFR, estimated glomerular filtration rate; ESRD , end-stage renal disease.


The ABC (age, biomarkers, clinical history) stroke risk score incorporates two biomarkers (NT-proBNP and high sensitivity cardiac troponin) and two clinical risk predictors (age and prior stroke) ( Fig. 15.7 ).




Fig. 15.7


The ABC (Age, Biomarkers, Clinical History) Stroke Risk Score.

Nomogram for the ABC risk score. For each predictor, read the points assigned on the 0–10 scale at the top and then sum these points. Find the number on the “Total Points” scale and then read the corresponding predictions of 1- and 3-year risk of stroke or systemic embolism below it. Continuous variables are represented from the 1st to the 99th percentiles. The prediction model is preferably used as a web-based calculator or app. NT-proBNP, N-terminal pro–B-type natriuretic peptide; SE, systemic embolism; TIA, transient ischemic attack.

(From Hijazi Z, Lindbäck J, Alexander JH, et al. The ABC (age, biomarkers, clinical history) stroke risk score: a biomarker-based risk score for predicting stroke in atrial fibrillation. Eur Heart J . 2016;37(20):1582–1590.)


It is important to note that all current risk scores for the prediction of ischemic stroke in AF perform modestly. The ACC/AHA/HRS and ESC guidelines recommend the CHA2DS2-VASc score for stroke risk stratification in AF.


Importantly, stroke risk assessment schemes have been established for patients with “nonvalvular” AF. Patients with mechanical valves require anticoagulation with vitamin K antagonists irrespective of the presence of AF. Also, AF patients with mitral stenosis are at a particularly high risk for systemic thromboembolism and have been excluded from any further trials studying anticoagulation regimens for AF. The 2014 AHA/ACC/HRS guidelines also include patients bioprosthetic heart valves or mitral valve repair in the definition of “valvular” AF.


On the other hand, stroke risk stratification using the above risk schemes appears to be adequate to guide treatment decisions regarding prophylactic anticoagulation therapy in patients with “nonvalvular” AF who have left valvular disease not included in the definition of “valvular” AF (e.g., nonrheumatic mitral regurgitation or aortic valve disease). In a recent report, left valvular disease was present in 22% of all nonvalvular AF patients, and although the embolic risk is higher in these patients compared with those without valve disease, neither the valve disease per se nor its severity was clearly associated with this risk, and a higher CHA2DS2-VASc score in these patients was likely to explain these results.


Bleeding Risk Stratification


Oral anticoagulation is associated with increased risk of bleeding. It is estimated that up to 44% of patients with AF have one or more absolute or relative contraindications for long-term oral anticoagulation therapy, most commonly related to increased risk of bleeding. Therefore an assessment of bleeding risk should be part of the patient assessment before starting anticoagulation. The risk of bleeding should be weighed against the potential benefit of stroke prevention in individual patients considered for anticoagulation therapy.


Several risk models have been proposed to predict bleeding risk on antithrombotic therapy. Only the HAS-BLED ( Table 15.9 ), HEMORR2HAGES ( Table 15.10 ), ATRIA ( Table 15.11 ), and ORBIT scores ( Table 15.12 ) have been derived or validated in AF populations.



TABLE 15.9

Clinical Characteristics Comprising the HAS-BLED Bleeding Risk Score








































Letter Clinical Characteristic Points
H Hypertension a 1
A Abnormal liver or renal function 1 or 2
S Stroke 1
B Bleeding 1
L Labile INR 1
E Elderly (age >65) 1
D Drugs or alcohol 1 or 2
Maximum score 9

INR, International normalized ratio.

a Hypertension is defined as systolic blood pressure >160 mm Hg. Abnormal kidney function is defined as the presence of long-term dialysis or renal transplantation or serum creatinine concentration of at least 200 µmol/L. Abnormal liver function is defined as chronic hepatic disease (e.g., cirrhosis) or biochemical evidence of significant hepatic derangement (e.g., bilirubin more than twice the upper limit of normal, in association with aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase more than three times the upper limit of normal). Bleeding refers to previous bleeding history or predisposition to bleeding, or both (e.g., bleeding diathesis, anemia). Labile INRs refers to unstable or high INRs or poor time in therapeutic range (e.g., <60%). Drugs or alcohol use refers to concomitant use of drugs (e.g., antiplatelet agents, nonsteroidal antiinflammatory drugs, or alcohol abuse).



TABLE 15.10

HEMORR2HAGES Bleeding Risk Score
























































Letter Clinical Characteristic Points
H Hepatic or renal disease 1
E Ethanol abuse 1
M Malignancy 1
O Older age 1
R Reduced platelet count or function 1
R Rebleeding risk 2
H Hypertension 1
A Anemia 1
G Genetic factors 1
E Excessive fall risk 1
S Stroke 1
Maximum score 12


TABLE 15.11

Anticoagulation and Risk Factors in Atrial Fibrillation Bleeding Risk Score

























Clinical Characteristic Points
Anemia 3
Severe renal disease 3
Age ≥75 years 2
Prior bleeding 1
Hypertension 1
Maximum score 10


TABLE 15.12

ORBIT Bleeding Risk Score































Letter Clinical Characteristic Points
O Older age (≥75 years old) 1
R Reduced hemoglobin, reduced hematocrit, or anemia 2
B Bleeding history 2
I Insufficient kidney function (eGFR <60 mg/dL per 1.73 m 2 ) 1
T Treatment with antiplatelets 1
Maximum score 7

eGFR, Estimated glomerular filtration rate.


In a study evaluating three bleeding risk scores (HAS-BLED, HEMORR2HAGES, and ATRIA), all three tested risk schemes demonstrated only modest performance in predicting the outcome of any clinically relevant bleeding, although the HAS-BLED score performed better than the HEMORR2HAGES and ATRIA scores; only HAS-BLED demonstrated a significant predictive performance for intracranial hemorrhage. Given its simplicity, the HAS-BLED score may be an attractive method for the estimation of oral anticoagulant–related bleeding risk for use in clinical practice, as recommended by the ESC guidelines. Patients are categorized as low, intermediate, and high bleeding risk according to HAS-BLED scores 0 to 1, 2, and 3 or higher, respectively. A score higher than 2 suggests a risk of major bleeding of 1.9% per year, whereas a score of 5 is associated with a risk of major bleeding of up to 12.5% per year.


It is important to understand that bleeding risk assessment is not a static phenomenon, and many common clinical factors that increase bleeding risk are potentially reversible. Further, a high bleeding risk score is not a reason to withhold anticoagulation therapy as such patients can potentially derive even greater net clinical benefit while on oral anticoagulation therapy. Instead, a high score should prompt careful review and follow-up as well as aggressive efforts at amelioration of potentially reversible bleeding risk factors (e.g., uncontrolled hypertension, labile international normalized ratios [INRs], balance problems, concomitant use of antiplatelet agents, alcohol excess, anemia, and renal or hepatic insufficiency).




Initial Evaluation


The initial evaluation of a patient with suspected or documented AF includes characterizing the pattern of the arrhythmia (e.g., paroxysmal or persistent), determining underlying causes (e.g., heart failure, pulmonary problems, hypertension, hyperthyroidism), defining associated cardiac and extracardiac conditions, and identifying potential complications of AF. In addition, a thorough history should be obtained to estimate of the risk of stroke (using the CHA2DS2-VASc scheme), bleeding risk, and quantify AF-related symptoms (e.g., CCS-SAF and modified EHRA scores). A careful history results in a well-planned focused work-up that serves as an effective guide to therapy.


The physical examination can suggest AF based on irregular pulse, irregular jugular venous pulsations, and variation in the intensity of the first heart sound. Examination can also disclose associated valvular heart disease, myocardial abnormalities, or heart failure.


Diagnostic Cardiac Testing


Ambulatory cardiac monitoring can be required for documentation of AF, its relation to symptoms, and evaluation of the adequacy of heart rate control. Transthoracic echocardiography is performed to evaluate for structural heart disease, assess cardiac function, and evaluate atrial size. Exercise testing is often used to assess the adequacy of rate control with exercise in permanent AF, to reproduce exercise-induced AF, and to evaluate for associated ischemic heart disease. Although ischemia per se does not appear to be a common cause of AF, identifying underlying coronary artery disease (in patients with risk factors) is particularly important if the use of a class IC antiarrhythmic drug is being considered.


Laboratory Testing


Laboratory evaluation includes assessment of serum electrolytes, renal, and hepatic function, and a blood count. Assessment for thyrotoxicosis is indicated for all patients with a first episode of AF, when the ventricular response to AF is difficult to control, or when AF recurs unexpectedly after cardioversion. Serum should be obtained for measurement of TSH and free thyroxine (T4), even if there are no other symptoms suggestive of hyperthyroidism, because the risk of AF is increased even in patients with subclinical hyperthyroidism.


Of note, plasma levels of B-type natriuretic peptide or N-terminal pro-B-type natriuretic peptide can be elevated in patients with paroxysmal and persistent AF in the absence of clinical heart failure, and levels decrease rapidly after restoration of sinus rhythm.


Electrophysiological Testing


Rarely, EP testing can be required, especially in patients with wide QRS complex tachycardia or a possible predisposing arrhythmia, such as AFL or paroxysmal SVT. Clues to the presence of paroxysmal SVT include a history of episodes of regular rapid palpitations dating from teenage or early adult years (unusual for AF to occur de novo in this age group) and termination of episodes of palpitations with vagal maneuvers or adenosine (which should not occur with AF).


Other Diagnostic Tests


Other diagnostics as guided by clinical presentation may include chest radiography, pulmonary function tests, and sleep study.


Screening for Atrial Fibrillation


AF is an important cause of embolic stroke, and subclinical (silent) AF should be considered in all survivors of an ischemic stroke, especially those of undetermined source (i.e., cryptogenic stoke). Studies have demonstrated that AF can be detected on ambulatory cardiac monitoring in more than 6% of unselected stroke patients and in up to 30% of those with cryptogenic stoke, with the latter accounting for approximately one-third of all ischemic strokes. In a recent meta-analysis, the overall proportion of newly detected AF was 7.4%, but varied widely depending on the timing, duration, and method of cardiac monitoring. Extending continuous electrocardiographic monitoring from 24 hours to 30 and 180 days increased the detection of AF from 4.2% to 15.2% and 29.2%, respectively. Hence, long-term (greater than or equal to 30 days) continuous ECG monitoring (noninvasive or using an implantable loop recorder) is recommended after cryptogenic stroke, and probably is also reasonable in all survivors of an ischemic stroke, even when another competing cause for stroke has been identified clinically (e.g., hypertension or carotid artery stenosis).


Opportunistic screening for silent AF (pulse palpation during a general practitioner consultation for any reason, followed by an ECG if the pulse was irregular) may also be considered in at-risk populations (e.g., individuals older than 65 years and patients with heart failure).


Furthermore, patients with pacemakers or defibrillators should be followed periodically and their devices interrogated on a regular basis for any evidence of AF, as suggested by automatic mode switch or atrial high-rate episodes. Wireless remote monitoring with predefined automatic alerts helps reduce the time to a clinical decision in response to the alert, compared with standard in-office follow-up.




Principles of Management


Management of AF should be aimed at identifying and treating underlying causes of the arrhythmia, as well as reducing symptoms, improving quality of life, and preventing cardiovascular morbidity and mortality associated with AF. There are four main issues that must be addressed in the treatment of AF: (1) prevention of systemic embolization; (2) ventricular rate control; (3) restoration and maintenance of NSR; and (4) risk factor modification.


The choice of therapy is influenced by patient preference, associated structural heart disease, severity of symptoms, and whether the AF is recurrent paroxysmal, recurrent persistent, or permanent. In addition, patient education is critical, given the potential morbidity associated with AF and its treatment. For control of symptoms, a safety-driven approach is of paramount importance because most treatments (drug, surgery, ablation) have the capacity to produce significant morbidity and even mortality.


Prevention of Systemic Embolization


Antithrombotic Drug Therapy


Antiplatelet therapy.


Aspirin is associated with only modest (22%) reduction in the incidence of stroke, corresponding to an absolute stroke risk reduction of 1.5% per year as compared with placebo. Thus except in the lowest risk patients, aspirin alone is not a viable treatment option for stroke prevention. The combination of aspirin plus clopidogrel is superior to aspirin therapy alone (28% relative risk reduction), but it is associated with a significantly increased risk of major bleeding (2.0% vs. 1.3% per year) to levels generally similar to those associated with warfarin therapy.


Vitamin K antagonists.


Vitamin K antagonists (warfarin) reduce stroke risk by approximately 64%, corresponding to an absolute annual strokes risk reduction of 2.7% as compared with placebo. Warfarin is superior to aspirin, with relative risk reduction of 39% for stroke and 29% for cardiovascular events. However, warfarin increases the risk of major bleeding by approximately 70% compared with aspirin. Although the risk of intracranial hemorrhage is doubled with adjusted-dose warfarin compared with aspirin, the absolute risk increase appears to be small (0.2% per year). In addition, randomized clinical trials have shown that warfarin is superior to the combination of aspirin plus clopidogrel for prevention of vascular events in patients with AF at high risk of stroke (relative risk reduction of 40%), with similar risks for major bleeding events. Combinations of warfarin (INR, 2.0 to 3.0) with antiplatelet therapy offer no incremental benefit in stroke risk reduction while they increase the risk of bleeding.


The reduction in ischemic stroke with warfarin in patients with paroxysmal AF is probably similar to that in patients with persistent or permanent AF. The benefit of warfarin is greatest for patients at higher risk of stroke, and there appears to be little benefit for those with no risk factors. The true efficacy of warfarin is likely to be even higher than suggested by trial results because many of the strokes in the warfarin-treated groups occurred in patients who were noncompliant at the time of the stroke.


The estimated annual incidence of bleeding associated with warfarin therapy is 0.6% for fatal bleeding, 3.0% for major bleeding, and 9.6% for major or minor bleeding. The risk of bleeding appears to be especially high during the first year of treatment. The addition of aspirin to warfarin further increases the rate of bleeding with a threefold increase in the rates of intracranial hemorrhage. Notably, the risk of major bleeding in older patients (greater than 80 years) receiving warfarin therapy, although higher than younger patients, is acceptably low (2.5% per year), and these patients can still benefit from warfarin prophylaxis when a good quality of anticoagulation is obtained. The risk of falling and intracranial bleeding should be considered but not overstated.


An INR between 2.0 and 3.0 is recommended for most patients with AF who receive warfarin therapy. The risk of stroke doubles when the INR falls to 1.7, and values up to 3.5 do not confer an increased risk of bleeding complications. A higher goal (INR between 2.5 and 3.5) is reasonable for patients at particularly high risk for embolization (e.g., prior thromboembolism, rheumatic heart disease, prosthetic heart valves). Similarly, in patients who sustain ischemic stroke or systemic embolism during treatment with therapeutic doses of warfarin (INR, 2.0 to 3.0), raising the intensity of anticoagulation to a higher INR range of 3.0 to 3.5 should be considered. This approach is probably preferable to adding an antiplatelet agent because an appreciable risk in major bleeding is seen with warfarin only when the INR is greater than 3.5 and is likely to be less than that associated with combination therapy.


Warfarin therapy is associated with several limitations that have dampened the enthusiasm of both patients and clinicians: a narrow therapeutic window that requires periodic INR monitoring and frequent dose adjustments, multiple drug and dietary interactions, genetic variability in response (accounting for 39% to 56% of the variability in the warfarin dose), long half-life (36 to 42 hours), and slow onset of action. In several trials, more than one-third of the patients refused warfarin therapy, largely because of the lifestyle changes required, the inconvenience of INR monitoring, and concern about bleeding risk. These issues have contributed to the underutilization of anticoagulation therapy in patients who can stand to derive benefit from it. In fact, it is estimated that less than 50% of eligible patients were treated with warfarin. Of those patients prescribed warfarin, there is ongoing attrition of its use to approximately 40% by 4 years.


Several series have highlighted the difficulties of maintaining the INR in the therapeutic range. More than one-third of patients taking warfarin are not maintained in the therapeutic range, thus exposing them to increased risk of either stroke (with subtherapeutic INRs) or bleeding (with supratherapeutic INRs). It has been found that if a patient’s INR is not maintained in the therapeutic range at least 65% of the time, the advantage of taking warfarin over aspirin is nullified.


Non–vitamin K antagonist oral anticoagulants.


There are two classes of non–vitamin K antagonist oral anticoagulants (NOACs): factor Xa inhibitors (such as rivaroxaban, apixaban, and edoxaban), and direct thrombin inhibitors (dabigatran). In general, NOACs are at least as effective as warfarin for the prevention of stroke and systemic thromboembolism in patients with nonvalvular AF and are much safer regarding the risk of intracranial hemorrhage compared to warfarin, with the annual rate ranging from 0.23% to 0.50%. However, limited data exist concerning the potential advantage of using NOACs in patients taking warfarin with optimal INR control (time in therapeutic range more than 75%).


NOACs have several potential advantages over warfarin, including their rapid onset of action, predictable therapeutic effect, less complex pharmacodynamics, limited dietary and drug interactions, and stable dose-related coagulation profile allowing for fixed dosing and obviating the need for routine monitoring. These advantages will likely promote greater use of anticoagulants, enhance patients’ compliance, allow for routine therapy without monitoring, and possibly eliminate the need for anticoagulation with parenteral agents such as heparin (“bridge therapy”). However, warfarin will remain the mainstay of treatment for patients with “valvular” AF and those with mechanical heart valves.


In a meta-analysis of trials randomizing NOACs to warfarin, NOACs were associated with: (1) significantly reduced composite stroke or systemic embolic events (19%), primarily driven by a reduction in hemorrhagic stroke; (2) a nonsignificant (14%) reduction in major bleeding, a reflection of decreased intracranial hemorrhage; and (3) a significant reduction in all-cause mortality. Major bleeding rates with these agents exceeded 2% to 3% per year, and minor bleeding rates were over 10% per year. In addition, by 2 years, 21% to 33% of patients discontinued the NOAC.


There are no direct head-to-head trials comparing the NOACs. In a meta-analysis using adjusted indirect comparisons, there was significant heterogeneity in results. Dabigatran lowered the composite of systemic emboli or stroke (vs. rivaroxaban), and apixaban lowered the risk of major gastrointestinal bleeding (vs. both rivaroxaban and dabigatran). Currently, there is no clear evidence to support a possible class-effect of a direct thrombin inhibitor or factor Xa inhibitors.


Nonpharmacological Interventions


The LAA has been implicated as the source of emboli in approximately 90% of patients with nonvalvular AF. Therefore several approaches have targeted exclusion of the LAA from the systemic circulation to prevent systemic thromboembolism and obviate the need for long-term oral anticoagulation therapy in patients with nonvalvular AF. These approaches can be of value in many AF patients, given the fact that up to 44% of patients with AF have one or more absolute or relative contraindications for chronic oral anticoagulation therapy, most commonly related to increased risk of bleeding. Furthermore, the safety and efficacy of chronic anticoagulation therapy can be limited by medication compliance, costs, and interactions with food and other medications.


Three main techniques are being utilized to accomplish LAA exclusion: percutaneous endocardial, percutaneous epicardial, and surgical approaches. Device-based endocardial LAA exclusion, such as Watchman (Boston Scientific, Natick, MA, United States) and Amplatzer Cardiac Plug (St. Jude Medical, Minneapolis, MN, United States), result in mechanical occlusion of the LAA. LAA exclusion with the endo-epicardial system (Lariat, SentreHEART, Redwood City, CA, United States) and surgical epicardial ligation result in both mechanical and electrical isolation of the LAA as a result of LAA infarction. Currently, these techniques are in different stages of evaluation and clinical development.


The ESC recommends percutaneous LAA closure device (Watchman and Amplatzer) use in nonvalvular AF patients with high stroke risk and contraindications to long-term oral anticoagulation. The 2014 AHA/ACC/HRS currently only recommends surgical excision of the LAA in patients undergoing cardiac surgery.


Potential candidates for percutaneous LAA closure include AF patients at high stroke risk with high risk of bleeding under oral anticoagulation, those with ischemic stroke despite well-controlled oral anticoagulation therapy, high probability of therapeutic noncompliance to oral anticoagulation, and intolerance to oral anticoagulation therapy due to severe hepatic or renal dysfunction or drug interactions ( Box 15.1 ). The procedure is contraindicated in patients at low stroke risk, those with valvular AF (e.g., mitral stenosis, mechanical cardiac valves), in the presence of other indications for long-term or lifelong oral anticoagulation therapy (e.g., venous thromboembolism, intracardiac clots), LAA thrombus, and contraindications for transseptal catheterization ( Box 15.2 ).



Box 15.1

Possible Indications for Percutaneous Left Atrial Appendage Closure in Patients With Nonvalvular Atrial Fibrillation





  • High stroke risk in conjunction to high bleeding risk under oral anticoagulation



  • Ischemic stroke despite well-controlled oral anticoagulation therapy



  • High probability of therapeutic noncompliance to oral anticoagulation



  • Intolerance to oral anticoagulation therapy due to severe hepatic or renal dysfunction or drug interactions




Box 15.2

Contraindications for Percutaneous LAA Closure in Patients With Nonvalvular AF





  • Low stroke risk



  • Valvular AF



  • Presence of other indications for long-term oral anticoagulation



  • LAA thrombus



  • Contraindications for transseptal catheterization



AF, Atrial fibrillation; LAA, left atrial appendage.



It is important to note that, although LAA exclusion procedures have been increasingly used for patients with nonvalvular AF, these procedures should not be considered universally as a substitute for oral anticoagulation therapy. Many of the disadvantages of warfarin therapy can be addressed by using NOACs rather than LAA exclusion, especially given the paucity of data supporting such procedures as compared to the large, prospective, randomized studies indicating the efficacy and safety of NOACs.


Recommendations for Long-Term Stroke Prevention


The cornerstone of management of patients with AF is adequate thromboprophylaxis. Essential to this is appropriate risk stratification and the need to balance the benefit of stroke prevention and the risk of bleeding with anticoagulant therapies (see above). Decision making for thromboprophylaxis by antithrombotic therapy must balance the risk of stroke against the risk of major bleeding, especially intracranial hemorrhage, which is associated with a high risk of death and disability.


Patients with valvular AF (those with mitral stenosis or valvular prosthesis) should be managed with oral anticoagulation. For nonvalvular AF, the CHA2DS2-VASc scoring system (see Tables 15.6 and 15.7 ) is currently the best validated and most clinically useful for risk stratification and is advocated by both European and United States guidelines. The initial decision step is to identify patients who are truly at low risk for ischemic stroke, in whom no antithrombotic therapy is recommended. These include patients without clinical stroke risk factors (i.e., CHA2DS2-VASc score of 0 in men or 1 in women); female gender does not appear to increase stroke risk in the absence of other stroke risk factors. On the other hand, oral anticoagulation therapy is recommended for patients of both sexes with CHA2DS2-VASc stroke risk score greater than or equal to 2. Those recommendations apply to all patients with AF irrespective of the type of AF (paroxysmal or nonparoxysmal).


There is uncertainty regarding the optimal antithrombotic therapy in low thromboembolic risk patients (i.e., CHA2DS2-VASc score of 1 in men or 2 in women). According to the 2014 AHA/ACC/HRS guidelines, no antithrombotic therapy or treatment with an oral anticoagulant or aspirin can be considered in these patients. On the other hand, recent studies, as well as ESC guidelines, support a positive advantage for stroke prevention with oral anticoagulation compared with no therapy or with aspirin in these patients. Furthermore, the use of NOACs may lower the threshold for initiating anticoagulation for AF patients, given the positive net clinical benefit of NOACs, even in patients with a CHA2DS2-VASc score of 1.


Importantly, treatment decisions should be individualized. Careful assessment of the risk of bleeding and patient preference is crucial. The expected clinical benefit of anticoagulation therapy should be balanced against the bleeding risk and should be thoroughly discussed with the informed patient. For equivocal cases, considering other possible risk predictors and risk models beyond the CHA2DS2-VASc scheme (e.g., renal function, biomarkers, findings on TEE) can potentially provide additional prognostic information and help identify those patients at a truly low thromboembolic risk.


The choice of anticoagulation therapy (warfarin vs. NOACs) is usually influenced by patient’s preference, comorbidities, renal function, cost, and drug interactions. The use of SAMe-TT2R2 score ( Table 15.13 ) can potentially identify those patients in whom warfarin therapy is more likely to be associated with labile INRs and, consequently, serious bleeding and thromboembolism (SAMe-TT2R2 score greater than 2). In those patients, NOACs are expected to offer a particular advantage.



TABLE 15.13

SAMe-TT2R2 score



































Letter Clinical Characteristic Points
S Sex (female) 1
A Age (<60 years) 1
Me Medical history a 1
T Treatment strategy (rhythm control) b 1
T Tobacco use (within 2 years) 2
R Race (nonwhite) 2
Maximum points 8

a Two of the following: hypertension, diabetes mellitus, coronary disease or prior myocardial infarction, peripheral vascular disease, congestive heart disease, previous stroke, pulmonary disease and hepatic or renal disease.


b Interacting drugs, e.g., amiodarone for rhythm control.



In high-risk patients who cannot be treated with oral anticoagulation because of poor tolerance or noncompliance issues or because of strong patient preference, dual antiplatelet therapy (aspirin plus clopidogrel) can be considered. However, dual antiplatelet therapy is not an alternative to oral anticoagulation in patients at high bleeding risk because the risk of major bleeding associated with dual antiplatelet therapy is generally similar to that with oral anticoagulation. In the latter group, aspirin monotherapy is associated with lesser bleeding risk, although at the expense of less protection from systemic thromboembolism. Percutaneous LAA exclusion procedures have become an important therapeutic alternative to long-term antithrombotic therapy in these patients.


Anticoagulation in the Pericardioversion Period


Patients without a contraindication to oral anticoagulation who have been in AF for more than 48 hours should receive 3 to 4 weeks of oral anticoagulation with warfarin or NOACs (with documented therapeutic INRs for those on warfarin) prior to and after cardioversion. This approach is also recommended for patients with AF who have valvular disease, evidence of LV dysfunction, recent thromboembolism, or AF of unknown duration.


The rationale for anticoagulation prior to cardioversion is based on observational studies showing that more than 85% of LA thrombi resolve after 4 weeks of anticoagulation therapy. Cardioversion-related clinical thromboembolic events have been reported in 5% to 7% of patients who did not receive anticoagulation before cardioversion (this risk appears to be much lower [less than 1%] for AF of less than 48-hour duration).


An alternative approach that eliminates the need for prolonged anticoagulation prior to cardioversion, particularly in low-risk patients who would benefit from earlier cardioversion, is the use of TEE-guided cardioversion. Cardioversion is performed if TEE excludes the presence of intracardiac clots. Anticoagulation after cardioversion, however, is still necessary.


After cardioversion, it is recommended to continue oral anticoagulation therapy for at least 4 weeks. This recommendation deals only with protection from embolic events related to the cardioversion period. Subsequently, the long-term recommendations for patients who have been cardioverted to NSR but are at high risk for thromboembolism are similar to those for patients with chronic AF, even though the patients are in NSR.


A different approach with respect to anticoagulation can be used in low-risk patients (with no mitral valve disease, severe LV dysfunction, or history of recent thromboembolism) in whom there is reasonable certainty that AF has been present for less than 48 hours. Such patients have a low risk of clinical thromboembolism (0.8% in one study) if converted early, even without surveillance TEE. The ACC/AHA guidelines do not recommend long-term anticoagulation prior to cardioversion in such patients, but they do recommend heparin use at presentation and during the pericardioversion period. The optimal therapy after cardioversion in this group is uncertain. A common practice is to administer aspirin for a first episode of AF that converts spontaneously and anticoagulation for at least 4 weeks in all other patients. Aspirin should not be considered for patients with AF of less than 48 hours’ duration if there is associated rheumatic mitral valve disease, severe LV dysfunction, or recent thromboembolism. Such patients should be treated the same as patients with AF of longer duration: 3 to 4 weeks of oral anticoagulation or shorter term anticoagulation with screening TEE prior to elective electrical or pharmacological cardioversion, followed by prolonged anticoagulation therapy after cardioversion.


Rate Control


Pharmacologic Therapy


Ventricular rate control during AF is important to prevent hemodynamic instability, improve symptoms and functional capacity, improve quality of life, and, over the long-term, prevent tachycardia-mediated cardiomyopathy. Oral or intravenous (IV) atrioventricular node (AVN) blockers are used for rate control, depending on the severity of symptoms and the degree of hemodynamic compromise caused by the tachycardia. In addition, correction of secondary causes of fast ventricular rates during AF (e.g., infection, hyperthyroidism, anemia, pain, and pulmonary embolism) is essential to achieve adequate rate control.


Beta-blockers or nondihydropyridine calcium channel blockers (verapamil and diltiazem) are the drugs of choice for rate control, and appear to have equivalent efficacy. Care should be used in administering those medications in patients with acutely decompensated heart failure. Beta-blockers are preferred in patients with cardiomyopathy, ischemic heart disease, and following surgical procedures. Verapamil and diltiazem are preferred in patients with reactive airway disease.


Digoxin is less effective and requires a longer time to achieve rate control, but may be considered if beta-blockers and calcium channel blockers have failed or have intolerable side effects. While digoxin reduces the resting heart rate, it is seldom effective in ambulatory patients because its effects are mediated by enhancement of vagal tone, which is offset during exertion. Thus digoxin has traditionally been used as a second-line agent, usually in sedentary patients or those with heart failure or hypotension. Recently, however, several systematic reviews and meta-analyses found that digoxin use was independently associated with a greater risk for mortality in patients with AF, regardless of concomitant heart failure. Some studies have suggested that AF nullifies the effect of digoxin in reducing hospitalizations for heart failure patients. Hence, the long-term use of digoxin is discouraged.


Amiodarone can be considered for rate control when other AVN blockers are unsuccessful or not tolerated. IV amiodarone is useful for acute control of the ventricular rate, and can be of particular value in acutely ill patients or those with acutely decompensated heart failure or severe hemodynamic compromise. Because of the probability of termination of AF by amiodarone, though very small, pericardioversion anticoagulation strategies should be considered, depending on the individual patient’s risk/benefit profile. Oral amiodarone can be useful for ventricular rate control when other measures are unsuccessful or contraindicated; however, long-term potential toxicity should be carefully considered.


In patients with AF and ventricular preexcitation causing rapid ventricular response, prompt direct-current cardioversion is recommended, especially when hemodynamic compromise is present. IV procainamide or ibutilide to restore NSR or slow the ventricular rate can be considered in hemodynamically stable patients. Importantly, drugs that preferentially slow AVN conduction without prolonging BT refractoriness (such as verapamil, diltiazem, adenosine, oral or IV digoxin, and IV amiodarone) can accelerate the ventricular rate and potentially precipitate hemodynamic collapse and VF in high-risk patients. Unlike the IV route of administration, chronic oral amiodarone therapy can slow or block BT conduction. Limited data exist regarding the use of beta-blockers; nonetheless, these drugs theoretically pose a similar potential risk and they should be used with caution.


Adequacy of rate control should be assessed at rest and with exertion. However, parameters for optimal rate control in AF remain controversial. It appears reasonable to target a resting heart rate of 60 to 80 beats/min and 90 to 115 beats/min during moderate exercise. Ambulatory monitoring can help assess adequacy of rate control; goals of therapy include a 24-hour average heart rate lower than 100 beats/min and no heart rate higher than 100% of the maximum age-adjusted predicted exercise heart rate. Also, a maximum heart rate of 110 beats/min during a 6-minute walk test is a commonly used target. Nonetheless, one study found that a more lenient rate control (resting heart rate less than 110 beats/min) is not inferior to strict rate control (heart rate less than 80 beats/min at rest and less than 110 beats/min during moderate exercise). Such an approach can be more convenient in clinical practice and can be considered especially in asymptomatic patients with permanent AF and no significant structural heart disease, but periodic monitoring of LV function is necessary to evaluate for the potential risk of tachycardia-mediated cardiomyopathy. Of note, lenient rate control does not seem to increase the risk of adverse atrial or ventricular remodeling.


In some patients with SND or tachycardia-bradycardia syndrome, pacemaker implantation can be required to protect from severe bradycardia while allowing the use of AVN blockers for adequate control of fast ventricular rates during AF or the use of antiarrhythmic drug therapy for maintenance of NSR ( see Fig. 8.6 ). In one report, nearly 20% of patients with AF required pacemaker placement for symptomatic bradycardia, most within 5 years of their AF diagnosis, which was especially common in those with a history of heart failure. In patients with SND, atrial or dual-chamber pacing significantly decreases the incidence of subsequent AF compared with ventricular pacing.


Atrioventricular Junction Ablation


Ablation of the AV junction combined with permanent pacemaker implantation (the “ablate and pace” approach), provides robust control of ventricular rate as well as regularization of the R-R interval. However, because it is permanent and mandates lifelong pacing, AV junction ablation usually is considered as a last resort approach in AF patients when rhythm control strategies fail and pharmacological rate control therapy is poorly tolerated or unsuccessful. AV junction ablation is especially useful when excessive ventricular rates induce a tachycardia-mediated decline in LV systolic function, despite appropriate medical therapy.


Furthermore, among patients with LV systolic dysfunction and AF, AV junction ablation has emerged as an important adjunctive therapy for cardiac resynchronization recipients. It has been estimated that 20% to 25% of those eligible for cardiac resynchronization have AF, and the cumulative incidence of new-onset AF/ATs ranges between 20% and 40% according to device interrogations. In patients with permanent AF or frequent persistent or paroxysmal arrhythmia episodes despite attempts to maintain NSR, intrinsic ventricular rate during AF (even though within the normal range) can override the biventricular pacing rate and reduce the percentage of effectively biventricular paced QRS complexes, thus precluding optimal ventricular resynchronization. Ablation of the AV junction in this setting has been associated with a reduction in all-cause mortality, a reduction in cardiovascular mortality, and an improvement in LVEF compared with those patients who were managed medically. It is important to note that the percentage of biventricular pacing determined by device counters often is artificially high because of invalid counting of fusion (hybrid between paced and intrinsic QRS morphologies) and pseudo–fusion complexes (pacing artifacts delivered but intrinsic QRS morphology not altered). In these patients, exercise ECG testing can help detect loss of effective ventricular synchronization and determine the percentage of pure biventricular pacing. AV junction ablation can also be required in ICD patients experiencing inappropriate therapies triggered by fast ventricular rates during AF.


Rhythm Control


Restoration and maintenance of NSR in patients with AF can have several potential benefits, including relief of symptoms, improved functional status and quality of life, and prevention of tachycardia-induced cardiomyopathy. Attenuation of electrical and structural atrial remodeling associated with AF (and hence retarding the progression of AF), and improvement in LV function also have been described. The impact of rhythm control on mortality, however, remains to be determined.


Unfortunately, complete maintenance of NSR (i.e., 100% freedom from AF recurrence) often is unachievable with current drug therapies and remains an impractical treatment goal. It has been estimated that the average 1-year recurrence rate associated with amiodarone approximates 35%, and the recurrence rates for other currently available antiarrhythmic drug therapies are even higher (more than 50%). However, it is likely that individuals with AF can derive benefit from even partial restoration of NSR.


Reversion to Normal Sinus Rhythm


When rhythm control strategy is chosen, both electrical and pharmacological cardioversion methods are appropriate options. The timing of attempted cardioversion is influenced by the duration of AF, severity of patient’s symptoms, adequacy of rate control, and risk of thromboembolism. Prompt cardioversion is recommended for patients with rapid ventricular rates and hemodynamic compromise attributed to AF (hypotension, acute heart failure, myocardial ischemia) or ventricular preexcitation, when rate-control drug therapy is unsuccessful or not tolerated. Cardioversion is also considered to restore NSR in stable but symptomatic patients with persistent AF, especially when ventricular rate control remains suboptimal.


Timing of cardioversion.


In stable patients with AF of a duration longer than 48 hours or of unknown duration, any mode of cardioversion (electrical, pharmacological, or ablation) should be delayed until the patient has been anticoagulated at appropriate levels for 3 to 4 weeks or TEE has excluded atrial thrombi, regardless of the CHA2DS2-VASc score. TEE can also be considered in patients with high thrombotic risk (e.g., severe valvular or congenital heart disease, prior thromboembolic events, severe cardiomyopathy), even when the duration of AF is less than 48 hours.


If urgency of cardioversion (because of severe symptoms or hemodynamic instability) precludes TEE, therapeutic doses of low-molecular-weight heparin, unfractionated heparin, or a non–vitamin K oral anticoagulant should be administered as soon as possible concurrent with or, preferably prior to, cardioversion, followed by long-term anticoagulation therapy.


Electrical cardioversion.


The overall success rate of electrical cardioversion for AF is about 90% and is inversely related to the duration of AF and LA size. The initial use of maximum-energy shocks, biphasic waveform, and anterior–posterior (as opposed to anterior–left lateral) electrode placement can help improve the efficacy of cardioversion and minimize the number of shocks required and, hence, the duration of sedation. In AF patients with ICDs, the electrodes should ideally be placed at least 8 cm away from the device in an anterior–posterior arrangement. The ICD can be used for internal cardioversion; however, the success rate is far lower than with external cardioversion, and each shock uses about 2 weeks of battery capacity.


Occasionally, electrical cardioversion fails to terminate AF, or AF recurs shortly after transient restoration of NSR. It is important to distinguish failure to terminate AF with a certain shock energy from successful termination of AF with nearly immediate recurrence. When AF fails to terminate, using higher energy levels, delivering a biphasic rather than monophasic waveform, applying external pressure on the cardioversion patches, changing the shock vector by altering the electrode pad position, and performing cardioversion during exhalation can improve effectiveness in some patients.


When AF recurs early after a successful cardioversion, repeated shocks at any energy are unlikely to have greater benefit. On the other hand, pretreatment with amiodarone, dofetilide, flecainide, ibutilide, propafenone, or sotalol can enhance success of electrical cardioversion and prevent early AF recurrence. In addition, pretreatment with a drug such as ibutilide can help lower the defibrillation threshold. The administration of IV magnesium sulfate alone before electric cardioversion does not appear to increase the rate of successful cardioversion of AF.


When the long-term use of antiarrhythmic drug therapy is planned for maintenance of NSR, initiation of drug therapy 1 to 3 days before electrical cardioversion (or a few weeks in the setting of amiodarone) to achieve effective drug levels at the time of cardioversion can help maintain NSR and prevent immediate recurrences of AF following cardioversion. This also confirms that the patient can tolerate the medication from a side effect perspective prior to cardioversion.


Of note, for AF of recent onset (less than 48 hours), newly started (less than 12 hours) AF episodes can be more difficult to terminate with electrical cardioversion than AF episodes lasting 12 to 48 hours, and failure of electrical cardioversion in the acute phase does not predict later successful cardioversion or spontaneous conversion to NSR in these patients.


Electrical cardioversion is usually preferred to pharmacological cardioversion because of greater efficacy and a low risk of proarrhythmia; however, it requires conscious sedation or anesthesia. Importantly, electrical cardioversion is contraindicated in patients with ongoing toxic reactions from digitalis or patients with hypokalemia.


The incidence of acute arrhythmic complications related to electrical cardioversion is very low. Ventricular arrhythmias needing intervention are extremely rare, irrespective of shock energy output or the concurrent use of antiarrhythmic drugs, although may be more common in patients receiving digitalis. Significant bradyarrhythmias (asystole greater than 5 seconds or heart rate less than 40 beats/min) resulting from SND and the effect of sedation are observed in about 1% of patients. Almost all external defibrillators have the capability of back-up bradycardia pacing through the defibrillation patches, which can be used transiently if needed. In addition, IV atropine or isoproterenol should be available. Of note, a large proportion (more than 40% in one report) of patients exhibiting severe bradyarrhythmias following successful cardioversion require pacemaker implantation during short-term follow-up.


Pharmacological cardioversion.


The efficacy of pharmacological cardioversion of AF is modest (30% to 70%), and is highest when initiated within 7 days of the onset of an episode of AF. While several antiarrhythmic drugs can be used for chemical cardioversion of AF, ibutilide and dofetilide are the most effective agents. Other antiarrhythmic drugs, including sotalol, amiodarone, and class IC agents (e.g., flecainide, propafenone) have limited efficacy. AVN blockers (beta-blockers, digoxin, and calcium-channel blockers) are generally not effective for restoration of NSR.


The risk of proarrhythmia is higher for chemical than electrical cardioversion. Therefore pharmacological cardioversion necessitates continuous cardiac monitoring (to detect SND, AV block, ventricular arrhythmias, and conversion into AF) for an interval that is dependent on the agent used (usually approximately half the drug elimination half-life).


Despite its limited efficacy, pharmacological cardioversion remains an option when sedation (which is required for electrical cardioversion) is not available or not well tolerated or when indicated by patient preference. In addition, as noted previously, when the use of long-term antiarrhythmic medications is planned for maintenance of NSR, starting drug therapy before electrical cardioversion can be beneficial, as it can help restore NSR in some patients and obviate the need for electrical cardioversion and, in other cases, can potentially enhance the efficacy of electrical cardioversion and prevent early recurrence of AF. Importantly, termination of AF may result in unanticipated sinus pauses/asystole with resultant presyncope or syncope, especially when using agents that can suppress sinus node function.


Ibutilide.


IV ibutilide can restore NSR in 28% to 51% of AF patients, with an average conversion time of less than 33 minutes. Pretreatment with ibutilide also improves the efficacy of electrical cardioversion. Importantly, ibutilide is associated with sustained polymorphic VT (torsades de pointes) in 1.2% to 2.4% of cases, and nonsustained VT in 1.8% to 6.7%, which is more likely to occur in patients with QT prolongation, marked hypokalemia, or a very low LVEF. Pretreatment with IV magnesium can increase the efficacy and reduce the risk of torsades de pointes.


Dofetilide.


Dofetilide can convert persistent AF to NSR in up to 60% of patients, typically within 36 hours of drug initiation. Dofetilide is rarely used solely for the purpose of cardioversion; rather, it is typically initiated for long-term rhythm control. When initiated in patients with persistent AF, electrical cardioversion is usually delayed for 24 to 48 hours to allow for potential pharmacological cardioversion. Of note, AF termination during dofetilide loading appears to be a predictor of durable response, even in longstanding persistent patients. Dofetilide is not available in Europe.


Amiodarone.


Amiodarone has very limited efficacy (approximately 25%) in terminating persistent AF, and is not a preferred agent solely for the purpose of cardioversion. When successful, conversion to NSR occurs several hours or days after initiation of IV amiodarone, and after days to weeks of long-term loading of oral amiodarone.


Flecainide and propafenone.


The class IC agents flecainide and propafenone can be used for pharmacologic cardioversion of AF; successful conversion to NSR typically occurs within 8 hours. The efficacy of flecainide for pharmacological cardioversion of recent-onset (less than 24 hours) AF is significantly higher than that of amiodarone, sotalol, procainamide, and propafenone, with conversion rates ranging between 52% and 92% in different reports. In comparison with oral flecainide, IV flecainide is no more effective for pharmacological cardioversion of recent-onset AF, although IV flecainide has a more rapid onset of action (mean time to cardioversion 55 vs. 110 minutes). Propafenone (IV or oral) can be used for the acute termination of AF (rate of conversion to NSR ranges from 56% to 83%). IV preparations of flecainide and propafenone are not available in the United States. Importantly, the use of class IC agents is contraindicated in patients with significant structural heart disease, particularly those with LV systolic dysfunction or coronary artery disease. In addition, class IC drugs can potentially convert AF into AFL with relatively slow atrial rate and, hence, facilitate 1 : 1 AV conduction and paradoxically faster ventricular rates. Therefore adequate rate control with AVN blockers (beta-blockers, diltiazem, verapamil) should be achieved before instituting antiarrhythmic therapy. Once the safety of pharmacological conversion with propafenone or flecainide has been established in the hospital setting, repeat patient-administered cardioversion using oral propafenone (450 to 600 mg) or flecainide (200 to 300 mg), in addition to a beta-blocker or nondihydropyridine calcium channel blocker, can be appropriate on an outpatient basis (the “pill-in-the-pocket” approach). This approach is usually employed in selected patients with infrequent symptomatic episodes of AF lasting at least several hours at a time and recurring less than once a month.


Vernakalant.


Vernakalant has been approved in Europe for the cardioversion of recent-onset AF (duration less than or equal to 7 days for patients not undergoing surgery, and less than or equal to 3 days for postcardiac surgery patients). IV vernakalant offers an AF conversion rate of about 62% within 90 minutes and appears to be much more effective than IV amiodarone. Some studies have also suggested its superiority to flecainide and propafenone. Vernakalant is not available in the United States.


Other antiarrhythmic agents.


Sotalol, dronedarone, quinidine, and procainamide offer very low efficacy for acute termination of AF, and are not recommended for pharmacological cardioversion.


Maintenance of Normal Sinus Rhythm


Only 20% to 30% of patients who are successfully cardioverted maintain NSR for more than 1 year without chronic antiarrhythmic therapy. This is more likely to occur in patients with AF for less than 1 year, no enlargement of the LA (less than 4.0 cm), and a reversible cause of AF (such as hyperthyroidism, pericarditis, pulmonary embolism, or cardiac surgery). It has been thought that the drugs most likely to maintain NSR suppress triggering ectopic beats and arrhythmias and affect atrial EP properties to diminish the likelihood of AF. There is therefore a strong rationale for antiarrhythmic drug therapy in patients who have a moderate to high risk of recurrence, provided that the therapy is effective and that toxic and proarrhythmic effects are low. Prophylactic drug treatment is seldom indicated in patients with a first-detected episode of AF and can also be avoided in patients with infrequent and well-tolerated paroxysmal AF.


Amiodarone has been directly compared to dronedarone, sotalol, and propafenone and found to be substantially more effective, with a 1-year rate of maintaining NSR of 65%. Dofetilide offers 50% to 65% efficacy in maintaining NSR at 1 year. Other antiarrhythmic agents have only modest efficacy (30% to 50% at 1 year). Drug selection is largely driven by the safety profile, the presence and extent of concomitant cardiovascular disease, hepatic and renal dysfunction, and drug-drug interactions. A safer, although possibly less efficacious, drug is usually recommended before resorting to more effective but less safe therapies ( Fig. 15.8 ; Table 15.14 ).




Fig. 15.8


Strategies for Rhythm Control in Patients With Paroxysmal and Persistent Atrial Fibrillation.

Drugs are listed alphabetically. a Catheter ablation is only recommended as first-line therapy for patients with paroxysmal AF (class IIa recommendation), depending on patient preference when performed in experienced centers. b Not recommended with severe LVH (wall thickness greater than 1.5 cm). c Should be used with caution in patients at risk for torsades de pointes ventricular tachycardia. d Should be combined with AV nodal blocking agents. AF, Atrial fibrillation; AV, atrioventricular; CAD, coronary artery disease; HF, heart failure; LVH, left ventricular hypertrophy.

(From January CT, Wann LS, Alpert JS, et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol . 2014;64:e1–e76.)


TABLE 15.14

Most Commonly Used Antiarrhythmic Drugs for the Treatment of Atrial Fibrillation




















































Antiarrhythmic Drug Year of Approval Channels Blocked Noncardiovascular Toxicity Cardiovascular Toxicity
Flecainide 1975 INa Dizziness, headache, visual blurring AFL with 1 : 1 conduction; VT; may unmask Brugada-type ST elevation
Propafenone 1976 INa, β-AR Metallic taste, dizziness, visual blurring AFL with 1 : 1 conduction; VT; may unmask Brugada-type ST elevation
Sotalol 1992 IKr, β-AR Bronchospasm Bradycardia, torsades de pointes
Amiodarone 1967 IKr, INa, ICaL, IKur, Ito, IKACh If, β-AR, α-AR Pulmonary (acute hypersensitivity pneumonitis, chronic interstitial infiltrates); hepatitis; thyroid (hypothyroidism or hyperthyroidism); photosensitivity; blue-gray skin discoloration; nausea; ataxia; tremor; alopecia Sinus bradycardia
Dronedarone 2009 IKr, INa, ICa, IKur, Ito, IKACh If, β-AR, α-AR Anorexia; nausea; hepatotoxicity Bradycardia
Dofetilide 2000 (US only) IKr None Torsades de pointes
Disopyramide 1962 INa, IKr, Acetylcholine Anticholinergic: dry mouth, urinary retention, constipation, blurry vision CHF exacerbation, torsades de pointes

AFL, Atrial flutter; AR, adrenoreceptor; CHF, congestive heart failure; ICaL, L-type Ca 2+ current; If, funny current; IKACh, acetylcholine-activated inward rectifier K + current; IKr, rapidly activating delayed rectifier K + current; IKur, ultra-rapidly activating delayed rectifier K + current; Ito, transient outward K + current; INa, Na + current; VT, ventricular tachycardia.


In patients with AF and minimal or no heart disease, flecainide, propafenone, sotalol, and dronedarone are preferred; amiodarone should be chosen later in the sequence of drug therapy because of its potential toxicity. In patients with adrenergically mediated AF, beta-blockers represent first-line treatment, followed by sotalol. The anticholinergic activity of long-acting disopyramide makes it a relatively attractive choice for patients with vagally mediated AF. In contrast, propafenone is not recommended in vagally mediated AF because its (weak) intrinsic beta-blocking activity may aggravate this type of paroxysmal AF.


In patients with substantial LV hypertrophy (LV wall thickness greater than 1.4 cm), it is recommended to avoid sotalol, flecainide, and propafenone because of concern of increased proarrhythmic risk. Dronedarone, although not specifically tested in this population, is likely to be safe. Amiodarone is usually considered when symptomatic AF recurrences continue to affect the quality of life in these patients.


In patients with coronary artery disease, sotalol, dofetilide, or dronedarone are recommended as first-line therapy, while flecainide and propafenone are contraindicated. Amiodarone is considered the drug of last resort in this population because of its potential toxicity.


Dofetilide and amiodarone are the only agents available for patients with AF with concomitant heart failure; other antiarrhythmic agents can be associated with substantial toxicity and proarrhythmia.


Quinidine is associated with increased mortality, likely the result of ventricular proarrhythmia secondary to QT interval prolongation. Hence, this drug has largely been abandoned for AF therapy.


Given the suboptimal efficacy of antiarrhythmic drug therapy, expectations and treatment goals have to be pragmatic. Reduction of the burden of AF and its impact on quality of life can be a reasonable outcome. Occasional AF recurrences may not require a change in antiarrhythmic drug therapy. When treatment with a single drug fails, combinations of antiarrhythmic drugs can be tried. Useful combinations include sotalol or amiodarone in addition to a class IC agent. However, when drug therapy is deemed unsuccessful and a rhythm control strategy is abandoned, antiarrhythmic drug should not be continued.


Amiodarone.


Amiodarone, although not approved by the US Food and Drug Administration for AF, is the most commonly prescribed and the most effective antiarrhythmic agent for the treatment of AF. However, the use of amiodarone is associated with significant adverse effects (including pulmonary, hepatic, thyroid, neurologic, and ophthalmic toxicity). QT prolongation is common but very rarely associated with torsades de pointes (0.5%). Although suppression of sinus and AV nodal function can occur early within the first few days of oral amiodarone therapy, the antiarrhythmic effect and QT prolongation can be delayed for days or weeks. A loading phase accelerates the onset of its antiarrhythmic activity. Amiodarone increases concentrations of warfarin, statins, and digoxin, and warfarin dose adjustment is often necessary. Appropriate periodic surveillance for lung, liver, and thyroid toxicity is required. Because of its potential toxicities, amiodarone should only be used after consideration of risks and when other agents have failed or are contraindicated.


Dofetilide.


Dofetilide offers up to 65% efficacy in maintaining NSR at 1 year. Dofetilide has been demonstrated to be reasonably safe in heart failure and post–MI populations. However, because of the risk of QT prolongation and VT, initiation of dofetilide requires a 3-day mandatory in-hospital loading period under continuous telemetry and ECG monitoring. Excessive QT prolongation or VT prompting drug discontinuation during the loading period has been reported in almost 20% of the patients. Concomitant usage of other QT-prolonging drugs increases the risk of these adverse events by almost twofold. Overall, the risk of torsades de pointes in patients receiving dofetilide ranges from 0.7% to 3.3%. In a retrospective cohort study of 1404 AF patients treated with dofetilide for a 5-year period, the incidence of torsades de pointes was 1.2%. Risk predictors included female gender, low LVEF, and greater QTc prolongation. Dofetilide is not approved in Europe.


Flecainide and propafenone.


Class IC agents are preferred first-line agents for rhythm control in patients with AF without structural heart disease, in whom both drugs are well tolerated and have a low risk of toxicity. On the other hand, these agents are contraindicated in patients with marked LV hypertrophy, coronary artery disease, or heart failure because of the risk of ventricular arrhythmias. Further, both flecainide and propafenone exhibit negative inotropic effects and should be avoided in patients with LV dysfunction. As noted previously, propafenone and flecainide are associated with a significant incidence of AFL with relatively slow atrial rate, which can be associated with 1 : 1 AV conduction and very fast ventricular rates; therefore adequate rate control with AVN blockers is recommended before instituting class IC drug therapy. In addition, class IC agents can delay His-Purkinje system (HPS) conduction and prolongation of the QRS duration, which when excessive (more than 25% compared with baseline) can be a marker for proarrhythmia risk.


Sotalol.


Sotalol has only modest efficacy in maintaining NSR (30% to 50% at 1 year). Sotalol causes drug-induced QT prolongation and torsades de pointes, especially in the setting of renal failure, hypokalemia, or the concomitant use of other QT-prolonging drugs. Therefore sotalol is often initiated in an inpatient setting with ECG monitoring to observe for excessive QT prolongation and proarrhythmia, especially when the drug is initiated during AF. However, drug initiation in an outpatient setting is also common, especially in low-risk patients with no underlying structural heart disease, QTc less than 450 milliseconds, normal electrolytes and renal function, and are in NSR at the time of drug initiation. Sotalol can be used in patients with ischemic heart disease, but should be avoided in patients with marked LV hypertrophy and those with renal insufficiency.


Dronedarone.


Dronedarone is a structural analogue of amiodarone that lacks the iodine moieties. Although dronedarone is associated with a lower incidence of noncardiovascular side effects than amiodarone, it is significantly less efficacious. The major cardiac adverse effects of dronedarone are bradycardia and QT prolongation. Torsades de pointes is rare but has been reported. Dronedarone can be used for AF in patients without structural heart disease, but is contraindicated (because of increased mortality) in patients with NYHA class III or IV heart failure and in patients who have had a recent (in the past 4 weeks) episode of decompensated heart failure, especially in the presence of LV systolic dysfunction. In patients with permanent AF, dronedarone increases the combined endpoint of stroke, cardiovascular death, and hospitalization. Therefore dronedarone is contraindicated in patients whose sinus rhythm is not restored.


Disopyramide.


Disopyramide is a sodium channel–blocking drug with potent anticholinergic and negative inotropic effects that can be considered for rhythm control in patients with AF. Because of its prominent vagolytic effects, disopyramide can be useful in “vagally mediated” AF (e.g., AF occurring in athletes or during sleep). Also, its negative inotropic effects make disopyramide beneficial in treating AF in patients with hypertrophic cardiomyopathy (HCM) associated with dynamic left ventricular outflow tract (LVOT) obstruction; however, these effects preclude its use in patients with underlying LV systolic dysfunction.


Rhythm Control Versus Rate Control


In the past, many physicians preferred rhythm control to rate control. Reversion of AF and maintenance of NSR restores normal hemodynamics and had been thought to reduce the frequency of embolism. However, two major randomized clinical trials—AFFIRM (Atrial Fibrillation Follow-Up Investigation of Rhythm Management) and RACE (RAte Control versus Electrical Cardioversion for Persistent Atrial Fibrillation)—compared rhythm and rate control in a select population at moderate stroke risk and found that embolic events occurred with equal frequency, regardless of whether a rate control or rhythm control strategy was pursued, and this occurred most often after warfarin had been stopped or when the INR was subtherapeutic. Both studies also showed an almost significant trend toward a lower incidence of the primary endpoint with rate control strategy. There was no difference in the functional status or quality of life. Hence, those trials provided evidence that both rhythm control and rate control are reasonable approaches (both strategies requiring long-term anticoagulation for stroke prevention), and suggested that rate control is an acceptable approach in most patients. The AF-Congestive Heart Failure (AF-CHF) study demonstrated similar results among patients with AF with concomitant heart failure. The results of those randomized controlled comparisons of rhythm and rate control therapies were confirmed by more recent observation studies, registries, and meta-analyses.


However, it would be incorrect to extrapolate that NSR offers no benefit over AF and that effective treatments to maintain NSR need not be pursued. First, these trials were not comparisons of NSR and AF; they compared a rate control strategy to a rhythm control strategy that attempted to maintain NSR but fell short, and crossover between treatment arms occurred at a high rate. The failure of AFFIRM and RACE trials in showing any difference between rate and rhythm control is not so much a positive statement for rate control but rather a testimony to the ineffectiveness of antiarrhythmic drug therapy in maintaining NSR over the long term. When the data from these trials were analyzed according to the patient’s actual rhythm (as opposed to his or her treatment strategy), the benefit of NSR over AF became apparent: the presence of NSR was found to be one of the most powerful independent predictors of survival, along with the use of warfarin, even after adjustment for all other relevant clinical variables. Patients in NSR are almost half as likely to die compared with those with AF. This benefit, however, is offset by the use of antiarrhythmic drug therapy, which increases the risk of death.


Therefore achieving and maintaining NSR remain viable and important treatment goals. However, because currently available antiarrhythmic agents commonly fail to suppress AF completely and have safety profiles that are less than ideal, it is reasonable to reserve it to the populations of patients likely to derive the greatest benefit from rhythm control. The selection of rhythm control or rate control strategies should be individualized and take into consideration the nature, intensity, and frequency of symptoms, patient preferences, comorbid conditions, and the risk of recurrent AF. According to analyses of available data, rhythm control can be an appropriate approach in young AF patients and those with newly diagnosed AF, significant symptoms, poorly controlled ventricular response, or tachycardia-mediated cardiomyopathy. On the other hand, asymptomatic or mildly symptomatic patients, especially those older than 65 years, and women with persistent AF who have hypertension or other underlying heart diseases can be better suited for rate control therapy.


It is important to note that current guidelines do not routinely recommend a rhythm control strategy for reducing the risk of mortality, stroke, or heart failure; rather, the primary indication for rhythm control therapy is for the reduction of symptoms and improvement in quality of life.


Catheter Ablation of Atrial Fibrillation


Catheter ablation of AF provides higher efficacy with comparable safety as antiarrhythmic drug therapy. AF ablation has been shown to significantly improve symptoms, exercise capacity, quality of life, and LV function, even in the presence of concurrent heart disease and when ventricular rate control has been adequate before ablation. Catheter ablation was also found to be associated with better quality of life, higher rates of freedom from both AF and antiarrhythmic medications, and lower rates of AF progression when compared with AVN ablation and biventricular pacing in symptomatic patients with AF and cardiomyopathy (LVEF less than or equal to 40%). Further, a recent study found that catheter ablation of AF superior to medical rate control strategy in patients with idiopathic cardiomyopathy, with significant improvement in the LVEF; restoration of NSR with catheter ablation resulted in significant improvements in LV systolic function, particularly in the absence of ventricular fibrosis on CMR. However, evidence is insufficient to determine whether AF ablation reduces all-cause mortality or stroke. Therefore the primary justification for an AF ablation procedure at this time is the presence of symptomatic AF.


At the current time, patient selection criteria for AF ablation should include weighing risks and potential benefits associated with the procedure, as well as consideration of other factors such as severity of symptoms, quality of life, presence and severity of structural heart disease and other comorbidities, and availability of other reasonable treatment options. In addition, the projected ablation success rate with the operator’s own experience and the tools available to him or her should be taken into consideration.


The ideal candidate for catheter ablation of AF has symptomatic episodes of paroxysmal or persistent AF, has not responded to one or more class I or III antiarrhythmic drugs, does not have severe comorbid conditions or severe structural heart disease, has an LA diameter smaller than 50 to 55 mm and, for those with longstanding AF, has had AF for less than 5 years. Catheter ablation of AF is likely to be of little or no benefit in patients with end-stage cardiomyopathy or massive enlargement of the LA (more than 60 mm), or in patients who have severe mitral regurgitation or stenosis and are deemed inappropriate candidates for valvular intervention. With improvements in the efficacy and safety of the procedure, the inclusion criteria for catheter ablation of AF continue to evolve; expanded indications at many centers now include patients with longstanding persistent AF and those with cardiomyopathy.


Current guidelines recommend catheter ablation in patients with symptomatic paroxysmal or nonparoxysmal AF as a second-line treatment after failure of or intolerance to class I or III antiarrhythmic drug therapy ( Fig. 15.9 ). It is reasonable to use similar indications for AF ablation in selected patients not well represented in clinical trials, including those with heart failure, cardiomyopathy, younger patients (less than 45 years) and older patients (greater than 75 years). However, the risks and benefits of catheter ablation must be carefully assessed in these patients. A lower success rate or a higher complication rate can be expected in AF patients with concomitant heart disease, obesity, sleep apnea, severe LA dilation, longstanding persistent AF, as well as frail, elderly patients.




Fig. 15.9


The 2017 HRS/EHRA/ECAS/APHRS/SOLAECE Indications for Catheter Ablation of Symptomatic Atrial Fibrillation (AF) .

Shown in this figure are the indications for catheter ablation of symptomatic paroxysmal, persistent, and longstanding persistent AF. The class for each indication based on whether ablation is performed after failure of antiarrhythmic drug therapy or as first-line therapy is shown.

(From Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation. Heart Rhythm . 2017;14:e275–e444.)


It is important to recognize that there are still no randomized controlled data demonstrating that a patient’s stroke risk is reduced by ablation. Therefore AF catheter ablation should not be performed with the sole intent of obviating the need for anticoagulation.


Complications of catheter ablation can have catastrophic outcomes in certain patients, including those with severe obstructive carotid artery disease, cardiomyopathy, aortic stenosis, nonrevascularized left main or three-vessel coronary artery disease, severe pulmonary arterial hypertension, or hypertrophic cardiomyopathy with severe LV outflow obstruction. Another relative contraindication is a history of major lung resection because of the severe impact of potential PV stenosis. Furthermore, because of the risk of thromboembolic events during the procedure and in the early postoperative period, patients who cannot be anticoagulated during and for at least 2 months after the ablation procedure should not be considered for catheter ablation of AF. Also, catheter ablation should not be performed in patients with an LAA thrombus or a recently implanted LAA closure device.


Catheter ablation as first-line therapy.


Recent studies have demonstrated superior efficacy of catheter ablation as a first-line therapy as compared to pharmacological therapy, though most patients enrolled in those studies were generally healthy with predominantly paroxysmal AF. According to the 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement, it is reasonable to consider catheter ablation as a first-line rhythm-control treatment (before therapeutic trials of class I or III antiarrhythmic drug therapy) for AF in select patients with symptomatic paroxysmal or persistent AF who prefer interventional therapy (see Fig. 15.9 ). This approach can be of particular value when poor tolerance to antiarrhythmic drugs is anticipated, such as in patients with tachycardia-bradycardia syndrome in whom pharmacological rhythm control strategies would necessitate pacemaker implantation. Similarly, catheter ablation is often recommended as an initial approach in high-level competitive athletes with paroxysmal or persistent AF in whom pharmacologic therapy can negatively affect athletic performance. The efficacy of this approach in unselected patient populations, however, still awaits confirmation by randomized studies, and the risk-benefit ratio of this approach in the individual patient should be carefully considered.


Catheter ablation for asymptomatic AF.


Symptoms of persistent AF can be quite subtle and nonspecific (e.g., lack of energy, effort intolerance) and may not be recognized by the patients or can be attributed to other comorbidities (such as sleep apnea or heart failure). Before labeling AF patients as “asymptomatic,” it is important to obtain a careful history to elicit symptoms. Also it is appropriate to attempt restoration of NSR (with electrical or pharmacological cardioversion, with or without long-term antiarrhythmic drug therapy) and then assess the patient’s symptom status while in NSR as compared to AF. Despite the lack of overt symptoms, AF ablation can still be a feasible way to improve well-being in these patients.


In addition, catheter ablation of AF may still be considered (class IIb) in select patients with truly asymptomatic paroxysmal or persistent AF (those who did not experience any improvement during a “trial of NSR”) when performed by an experienced operator and following a detailed discussion of the risks and benefits. The patient should be informed that AF, whether symptomatic or asymptomatic, is associated with an increased risk of stroke, heart failure, dementia, and mortality, and while it is possible that maintenance of NSR with AF ablation can potentially reduce these risks, these potential benefits remain unproven. Deferring ablation while awaiting the results of clinical trials can potentially allow progression of AF to a stage when the efficacy of AF ablation is significantly reduced. Importantly, while this approach can be acceptable in select asymptomatic patients, it is not recommended for patients with longstanding persistent AF and those with clinical whose clinical profile would be associated with a low procedural efficacy and safety.


Surgical Ablation of Atrial Fibrillation


The classic Cox-maze procedure involves creating a series of incisions in the left and right atria designed to direct the propagation of the sinus impulse through both atria while interrupting the multiple macroreentrant circuits thought to be responsible for AF. This procedure is the most effective means of curing AF, eliminating the arrhythmia in 75% to 95% up to 15 years after surgery. Improvements and simplifications of the surgical technique culminated in the Cox-maze III procedure, which became the gold standard for the surgical treatment of AF. Nonetheless, because of its complexity, technical difficulty, and risk of mortality and other complications, the maze procedure did not gain widespread acceptance.


To simplify the procedure, the standard cut-and-sew surgical technique has been replaced with linear epicardial ablation using unipolar or bipolar RF ablation, cryoablation, laser, high-frequency ultrasound, or microwave energy. Most surgical epicardial ablation procedures have been performed in conjunction with mitral valve surgery; the combination of mitral valve repair and cure of AF can enable selected patients to avoid life-long anticoagulation.


Current surgical instrumentation now enables minimally invasive approaches to be performed epicardially on the beating heart through mini-thoracotomies with video assistance. Bipolar RF is the predominant energy source used, and bilateral PV isolation is the most common lesion set, with some approaches adding ganglionic plexus ablation, as well as exclusion of the LAA. However, despite elimination of the need for median sternotomy and cardiopulmonary bypass, these procedures are still relatively invasive. To minimize the invasiveness of the procedure further, a totally thoracoscopic approach has been developed. Although multiple series described high success rates for paroxysmal AF (89% at 12 months of follow-up), success has been limited in patients with persistent and longstanding persistent AF (25% to 87%). In one report, the overall complication rate was 10%, with a perioperative mortality rate of 1.8%.


More extensive ablation lines, in addition to PV antral isolation and ganglionic plexus ablation, as well as documentation of complete PV isolation by demonstration of EP entrance or exit block, conduction block across ablation lines, and the detection and confirmation of ablation of the parasympathetic component of the ganglionic plexuses, are being evaluated to improve outcome. Some series reported a single procedure success rate of 86% at 1 year without the use of antiarrhythmic drugs.


Surgical ablation of AF is recommended in patients undergoing concomitant open heart surgery (whether ablation is performed after failure of antiarrhythmic drug therapy or as first-line therapy and regardless of the duration of the arrhythmia). Such an approach is also reasonable in patients undergoing closed cardiac surgery (e.g., coronary bypass or surgery) ( Fig. 15.10 ).




Fig. 15.10


The 2017 HRS/EHRA/ECAS/APHRS/SOLAECE Indications for Surgical Ablation of Atrial Fibrillation (AF) .

Shown in this figure are the indications for surgical ablation of paroxysmal, persistent, and longstanding persistent AF. The class for each indication based on whether ablation is performed after failure of antiarrhythmic drug therapy or as first-line therapy is shown. The indications for surgical AF ablation are divided into whether the AF ablation procedure is performed concomitantly with an open surgical procedure (such as mitral valve replacement), a closed surgical procedure (such as coronary artery bypass graft [CABG] surgery), or as a stand-alone surgical AF ablation procedure performed solely for treatment of AF. AVR, Aortic valve replacement.

(From Calkins H, Hindricks G, Cappato R, et al. 2017 HRS/EHRA/ECAS/APHRS/SOLAECE expert consensus statement on catheter and surgical ablation of atrial fibrillation. Heart Rhythm . 2017;14:e275–e444.)


Currently, stand-alone and hybrid surgical AF ablation can be considered for symptomatic patients with AF who were refractory to one or more attempts at catheter ablation or who are not candidates for catheter ablation (e.g., patients not candidates for long-term anticoagulation and those with an LA thrombus). Although surgical ablation was found in one report to have superior efficacy to catheter ablation, the complication rate after surgical ablation was higher. Hence, the decision to recommend surgical AF ablation before considering catheter ablation for patients with symptomatic AF refractory to drug therapy and no other indication for cardiac surgery remains controversial, and it should be based on institutional experience with both techniques, the relative outcomes and risks of each in the individual patient, as well as patient preference (see Fig. 15.10 ). Given the degree of patient discomfort, longer hospitalizations and recovery times, and the risk of bleeding following surgery, most patients prefer catheter to surgical ablation.


Other Nonpharmacological Approaches for Rhythm Control


Atrial antitachycardia pacing.


Several pacing algorithms have been developed to inhibit the initiation of AF episodes. These include continuous atrial pacing just faster than the intrinsic sinus rate, overdrive atrial pacing after a PAC, algorithms for preventing pauses after PACs, and overdrive atrial pacing after the termination of an episode of AF to suppress an early arrhythmia recurrence. A multitude of studies tested the efficacy of those algorithms and showed no consistent reduction in AF burden or improvement in AF symptoms. Similarly, dual-site atrial pacing and atrial septal pacing site did not show benefits.


Modern pacemakers are equipped with a variety of atrial antitachycardia pacing (ATP) algorithms designed to terminate atrial tachyarrhythmias. ATP is delivered at an atrial CL shorter than the detected arrhythmia with the administration of a number of pulses of fixed duration (burst pacing), or sequences at progressively shorter intervals (ramp pacing) to abort episodes of AFL or AT. Prior generations of ATP algorithms demonstrated a modest efficacy (30% to 60%) in terminating slow regular ATs, less effective with rapid ATs, and ineffective at treating established AF. Further, the clinical impact of these algorithms on the burden of the arrhythmia is small. It is important to note that, besides the need for organized atrial tachyarrhythmias, the efficacy of ATP is crucially reliant on early detection of AT and correct rhythm classification by the device.


More recently, a new-generation atrial ATP (“reactive ATP”) was developed to target atrial tachyarrhythmias at onset (when the atrial CL is relatively long) and after any change in rate or regularity when the episode may be most amenable to termination by pacing. Transitions toward more regular or slower rhythms are not infrequent (reportedly occurring in 64% of atrial tachyarrhythmia episodes), even after hours following the onset of the arrhythmia. Unlike standard ATP algorithms, reactive ATP continues to monitor atrial rhythm and watches for any change in rate or regularity, and then opportunistically applies ATP when the episode is most amenable to termination by pacing ( eFig. 15.5 ). In a recent study, reactive ATP reduced the risk of progression of AF to permanent or persistent AF.





eFig. 15.5


Examples of Atrial Antitachycardia Pacing (ATP) .

Upper panel, Ramp ATP therapy delivered during atrial fibrillation fails at terminating the arrhythmia. Lower panel, In the same patient, ramp ATP therapy delivered when the tachycardia transitions into a more organized and slower arrhythmia successfully terminates the tachycardia and restores atrial-paced rhythm within 2 seconds of ATP delivery. AF, Atrial fibrillation; AP, atrial-paced event; AT, atrial tachycardia; TD, tachycardia detection; TP, tachycardia pacing; TS, tachycardia-sensed; VP, ventricular-paced event; VS, ventricular-sensed event.


Currently, pacemaker implantation is not indicated for the sole purpose of prevention or treatment of AF in patients without other indications for pacemaker implantation. Nonetheless, ATP offers a therapeutic option for rhythm control in patients with permanent dual-chamber pacemakers or defibrillators that have this feature.


Atrial defibrillators.


Atrial defibrillators can terminate AF with high acute success rates, but the need for repeated shocks and the resulting patient discomfort often render this option intolerable. Therefore implanted defibrillators are no longer recommended for rhythm control in AF patients.


Upstream Therapy


Upstream therapy refers to the use of non-ion-channel drug therapy that modifies the atrial substrate upstream of AF to reduce susceptibility to, or progression of, AF. The goal of this approach is attenuation and reversal of atrial structural remodeling to prevent new-onset AF (i.e., primary prevention) or recurrent AF (i.e., secondary prevention). Although some studies demonstrated a potential value of upstream therapy for primary prevention of AF in selected patients, data regarding its value for secondary prevention have been disappointing.


Given the role of fibrosis and inflammation in the pathogenesis of AF, drugs that suppress fibrosis, as well as antiinflammatory and antioxidative drugs, are being investigated both alone and in combination with traditional antiarrhythmic drug therapy. Among these drugs are several angiotensin-converting enzyme inhibitors, angiotensin II type 1 receptor blockers, antialdosterone agents, statins (3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors), and omega-3 polyunsaturated fatty acids. These agents seem to reduce atrial fibrosis and were found to potentially reduce atrial structural remodeling and AF susceptibility in various AF experimental models. However, clinical trials produced inconclusive results, both for the primary and secondary prevention of AF.


Although several studies have suggested a beneficial effect of renin–angiotensin–aldosterone system inhibitors for primary prevention of AF in patients with heart failure and LV systolic dysfunction or hypertrophy, no convincing benefit has been observed in patients without underlying heart disease or for secondary AF prevention. Therefore inhibitors of the angiotensin axis can be considered for AF management when the arrhythmia is associated with other underlying conditions that are themselves associated with myocardial fibrotic remodeling (e.g., LV systolic dysfunction and possibly hypertension with LV hypertrophy), but are not recommended in patients with no apparent cardiovascular disease.


No convincing evidence currently exists to support the use of polyunsaturated fatty acids or fish oil for either primary or secondary AF prevention. While some studies demonstrated a protective effect of statins against new-onset AF in patients undergoing coronary artery bypass graft surgery, other studies arrived at conflicting results. On the other hand, short-term colchicine has been associated with lower rates of postoperative AF and reduced early AF recurrence after catheter ablation.


Risk Factor Management


There is growing evidence supporting aggressive risk factor modification for primary prevention of AF, management of symptomatic AF, reducing the risk of AF recurrence postablation, and reducing thromboembolic complications of AF ( Fig. 15.11 ). Pursuing and managing hypertension, diabetes, sleep apnea, obesity, and alcohol consumption need to be adopted as a systematic routine in the management of AF patients.




Fig. 15.11


Risk Factor Management for Prevention of Atrial Fibrillation.

ACEI, Angiotensin converting enzyme inhibitor; AHI, apnea–hypopnea index; ARB, angiotensin receptor blocker; BMI, Body mass index; BP, blood pressure; CPAP, continuous positive airway pressure; LDL, low-density lipoprotein; TG, triglyceride.

(From Lau DH, Schotten U, Mahajan R, et al. Novel mechanisms in the pathogenesis of atrial fibrillation: practical applications. Eur Heart J . 2016;37:1573–1581.)


Hypertension


Hypertension is an independent risk factor for AF and stroke. Although treatment of hypertension has not been consistently shown to decrease AF risk, it is an important component of reducing cardiovascular complications and thromboembolic risk.


Diabetes


Although diabetes is an independent risk factor for the development of AF and thromboembolic complications, there are limited data on diabetes management and AF risk. Intense glycemic control does not appear to directly impact the incidence or course of AF. Nonetheless, optimization of diabetes management and prevention of cardiovascular complications may indirectly reduce risk of AF.


Sleep Disordered Breathing


An independent association exists between AF and obstructive sleep apnea. Therefore AF patients should be actively screened for undiagnosed obstructive sleep apnea and evaluated when clinically suspected. Treatment of sleep apnea, when diagnosed, is an important component of AF management. CPAP therapy is associated with more than 40% relative risk reduction in AF recurrence in patients with obstructive sleep apnea, regardless of the AF treatment strategy (pharmacological or invasive).


Lifestyle Modifications


Lifestyle interventions aimed at maintaining a healthy body weight and cardiorespiratory fitness, in the context of a comprehensive risk factor modification, are recommend for both prevention and management of AF. Weight reduction in overweight and obese AF patients, especially those with type II diabetes and hypertension, can improve management of concomitant cardiometabolic risk factors, and was shown to improve symptoms and reduce AF burden.


In addition, regular, moderate exercise appears to benefit overweight and obese patients with symptomatic paroxysmal or persistent AF. One study found a significant dose-response relationship between baseline cardiorespiratory fitness with a 20% reduction in the risk of AF recurrence for each metabolic equivalent (MET) increase in baseline cardiorespiratory fitness. The benefits gained from cardiorespiratory fitness are additive to the effect of weight loss. Increased cardiorespiratory fitness is associated with beneficial effects on blood pressure, diabetic control, lipid profile, and inflammation, all of which can potentially contribute to reduced AF burden. However, guidelines on appropriate weight loss and fitness targets are still lacking.


Habitual and binge alcohol drinking increases the risk of new-onset AF as well as the risk of arrhythmia progression in patients with AF. Therefore reducing alcohol consumption can potentially be an effective strategy for primary and secondary AF prevention. In this context, however, a safe level of daily alcohol consumption in AF patients has not been established. In addition, appropriate counseling should be provided for smoking cessation and recreational drug abuse.


Management of Postoperative Atrial Fibrillation


Primary Prevention


Multiple strategies for preventing postoperative AF have been studied. Prophylaxis of AF using perioperative beta-blockers, amiodarone, and sotalol has shown promising results. However, no strategy completely eliminates the occurrence of postoperative AF.


Beta-blockers.


Oral beta-blockers have been consistently shown to reduce the development of postoperative AF (from 39% to 31%, in one report) and, in the absence of contraindications, are strongly recommended for virtually all patients undergoing cardiac surgery. Generally, oral beta-blocker therapy is started at least 2 to 3 days before surgery, or within 24 hours after surgery if not given preoperatively. The dose is up-titrated as tolerated. In patients already receiving chronic beta-blocker therapy, the drug should be continued without interruption perioperatively.


Amiodarone.


Amiodarone reduces the incidence of postoperative AF by more than 50% (compared with placebo); however, its incremental value is less well defined when compared with beta-blocker therapy. Preoperative use of amiodarone to prevent postoperative AF is a class IIa recommendation in the 2014 ACC/AHA/HRS guidelines.


Different regimens of amiodarone have been evaluated. Oral regimens were started at 1, 5, or 7 days before nonemergent surgery and continued for several days postoperatively. IV regimens involve starting amiodarone infusion immediately before or immediately after surgery, which is continued for 48 hours followed by oral therapy for 3 to 4 days. In a recent study, the efficacy of amiodarone in prevention of postoperative AF was maintained irrespective of route (oral vs. IV) and timing of administration (preoperative vs. immediate postoperative), and regardless of the duration of therapy, when at least 300 mg of IV amiodarone was loaded, and a total dose of 1 g was administered.


Sotalol.


Sotalol was found to be more effective than beta-blockers for postoperative AF prophylaxis. There was no significant difference in the rates between sotalol and amiodarone. Sotalol therapy is usually started 24 to 48 hours before surgery or four hours after surgery. However, the risk of bradycardia and torsade de pointes, especially in those with electrolyte disturbances, has limited the widespread use of sotalol for the prevention of postoperative AF.


Other therapies.


Despite some suggestion of beneficial effects, current evidence does not support the routine prophylactic use of corticosteroids, atrial pacing, posterior pericardiotomy, antioxidant vitamins C and E, n-3 polyunsaturated fatty acids, statins, magnesium, or colchicine to prevent postoperative AF in the cardiac surgical population. Studies using verapamil, digoxin, or procainamide showed no significant benefits compared to a placebo.


Perioperative infusion of human natriuretic peptide (carperitide), which inhibits the renin–angiotensin–aldosterone system, was recently shown to reduce the occurrence of postoperative AF in patients undergoing coronary bypass grafting.


Of note, prophylactic PV epicardial isolation in patients undergoing coronary bypass grafting does not decrease the incidence of postoperative AF or its clinical impact.


Rate Versus Rhythm Control


Beta-blockers are the drugs of choice for rate control in patients with postoperative AF. The indications for cardioversion of AF and recommendations for rhythm- versus rate-control strategies are similar to those discussed for nonsurgical patients. When antiarrhythmic drug therapy is required for rhythm control, amiodarone is the drug of choice. Sotalol can be considered if amiodarone is contraindicated. Class IC agents, such as flecainide and propafenone, generally are avoided in these patients given the presence of structural heart disease.


Prevention of Systemic Embolization


Guidelines for stroke prevention in nonsurgical AF patients apply to those with postoperative AF. Most of these patients have multiple stroke risk factors but also increased risk of bleeding in the postoperative phase. Therefore the decision to initiate anticoagulation therapy should be guided by the individual patient’s bleeding risk and CHA2DS2-VASc score. Data are lacking regarding the threshold burden or duration of postoperative AF at which anticoagulation is favored, but AF lasting for longer than 48 hours should prompt strong consideration of anticoagulation therapy. The optimal duration for which anticoagulation must be continued after cessation of postoperative AF is uncertain.


Follow-Up of Patients With Postoperative Atrial Fibrillation


Follow-up is recommended at 6 to 12 weeks after surgery to evaluate the presence of persistent or paroxysmal AF and reassess management strategy. Ambulatory cardiac monitoring should be considered to screen for asymptomatic paroxysmal arrhythmias. The optimal frequency and intensity of cardiac rhythm monitoring beyond the 3-month period are not known. If AF is documented, long-term anticoagulation should be considered based on the CHA2DS2-VASc score, and the need for rate versus rhythm control should be reassessed. If there is no evidence of symptomatic or asymptomatic AF beyond the immediate postoperative period, discontinuation of antiarrhythmic drug therapy, if initiated after cardiac surgery, is recommended.




Electrocardiographic Features


Atrial Activity


AF is characterized by rapid and irregular atrial fibrillatory waves (f waves) and a lack of clearly defined P waves, with an undulating baseline that can alternate between recognizable atrial activity and a nearly flat line ( Fig. 15.12 ). Atrial fibrillatory activity is generally best seen in lead V 1 and in the inferior leads. Less often, the f waves are most prominent in leads I and aVL.




Fig. 15.12


Surface Electrocardiogram of Atrial Fibrillation With Chronic Right Bundle Branch Block.


The rate of the fibrillatory waves is generally between 350 and 600 beats/min. With up to 600 impulses generated every minute, syncytial contraction of the atria is replaced by irregular atrial twitches. Therefore the fibrillating atria look like a bag of worms in that the contractions are very rapid and irregular. The f waves vary in amplitude, morphology, and intervals, thus reflecting the multiple potential types of atrial activation that may be present at the same time at different locations throughout the atria. The f waves can be fine (amplitude less than 0.5 mm on ECG) or coarse (amplitude more than 0.5 mm). On occasion, the f waves can be inapparent on the standard and precordial leads, which is most likely to occur in permanent AF. It was initially thought that the amplitude of the f waves correlated with increasing atrial size; however, echocardiographic studies have failed to show a correlation among the amplitude of the f waves, the size of the atria, and the type of heart disease. The amplitude, however, may correlate with the duration of AF.


AF should be distinguished from other rhythms in which the R-R intervals are irregularly irregular. These include multifocal AT (see Fig. 15.2 ), wandering atrial pacemaker, multifocal PACs, and AT or AFL with varying AV block. In general, distinct (although often abnormal and possibly variable) P (or flutter) waves are present during these arrhythmias, in contrast to AF. Patients with rheumatic mitral stenosis often demonstrate large-amplitude fibrillatory waves in the anterior precordial leads (V 1 and V 2 ), which can be confused with AFL. However, careful examination of the fibrillatory waves reveals them to have a varying CL and morphology. The distinction between AF and AFL can also be confusing in patients who demonstrate a transition between these arrhythmias. Thus AF may organize to AFL or AFL may degenerate to AF ( eFig. 15.6 ). Occasionally, extracardiac artifacts (e.g., 60 cycle/min muscle tremors, as in parkinsonism) can mimic f waves.





eFig. 15.6


Surface Electrocardiogram and Intracardiac Recordings Demonstrating Spontaneous Conversion of Atrial Fibrillation (At Left) Into Typical Atrial Flutter (Right) .

CS dist , Distal coronary sinus; CS prox , proximal coronary sinus; HB, His bundle; HRA , high right atrium.


Atrioventricular Conduction During Atrial Fibrillation


The ventricular response in AF is typically irregularly irregular, and the ventricular rate depends on multiple factors, including the EP properties of the AVN, the rate and organization of atrial inputs to the AVN, the level of autonomic tone, the effects of medications that act on the AV conduction system, and the presence of preexcitation over an AV BT.


The ventricular rate in untreated patients usually ranges from 90 up to 170 beats/min. Ventricular rates that are clearly outside this range suggest some concurrent influence. Ventricular rates slower than 60 beats/min are seen with AVN disease and can be associated with the sick sinus syndrome, drugs that affect conduction, and high vagal tone, as can occur in a well-conditioned athlete. The ventricular rate in AF can become rapid (more than 200 beats/min) during exercise, with catecholamine excess ( Fig. 15.13 ), parasympathetic withdrawal, thyrotoxicosis, or preexcitation ( Fig. 15.14 ). The ventricular rate can be very rapid (more than 300 beats/min) in patients with the Wolff-Parkinson-White syndrome, with conduction over AV BTs having short anterograde refractory periods.




Fig. 15.13


Surface Electrocardiogram of Atrial Fibrillation With Rapid Ventricular Response in a Patient With Septic Shock on Dopamine Infusion.



Fig. 15.14


Preexcited Atrial Fibrillation (AF) .

(A) Electrocardiogram (ECG) showing normal sinus rhythm (NSR) with a Wolff-Parkinson-White pattern and preexcitation using a left lateral bypass tract (BT). (B) ECG showing preexcited AF (i.e., AF with conduction over the BT).


The compact AVN is located anteriorly in the triangle of Koch. There are two distinct atrial inputs to the AVN, anteriorly via the interatrial septum and posteriorly via the crista terminalis ( see Chapter 9 ). Experiments in a rabbit AVN preparation demonstrated that propagation of impulses during AF through the AVN to the His bundle (HB) is critically dependent on the relative timing of activation of septal inputs to the AVN at the crista terminalis and interatrial septum. Other investigators showed that the ventricular response also depends on atrial input frequency.


Concealed conduction likely plays the predominant role in determining the ventricular response during AF. The constant bombardment of atrial impulses into the AVN creates substantial and varying degrees of concealed conduction, with atrial impulses that enter the AVN but do not conduct to the ventricle, thus leaving a wake of refractoriness encountered by subsequent impulses. This also accounts for the irregular ventricular response during AF. Although the AVN would be expected to conduct whenever it recovers excitability after the last conducted atrial impulse, which would then be at regular intervals, the ventricular response is irregularly irregular because of the varying depth of penetration of the numerous fibrillatory impulses approaching the AVN, leaving it refractory in the face of subsequent atrial impulses.


Alterations of autonomic tone can have profound effects on AVN conduction. Enhanced parasympathetic and sympathetic tone have negative and positive dromotropic effects, respectively, on AVN conduction and refractoriness. An additional factor is the use of AVN blocking agents such as digoxin, calcium channel blockers, or beta-blockers. There also may be a circadian rhythm for both AVN refractoriness and concealed conduction that accounts for the circadian variation in ventricular rate.


Ventricular Preexcitation During Atrial Fibrillation


The presence of a grossly irregular, very rapid ventricular response (more than 250 beats/min) with QRS duration longer than 120 milliseconds during AF rarely results from conduction over the AVN and strongly implies conduction over an AV BT (see Fig. 15.14 ). At very fast heart rates, there is usually a tendency toward regularization of the R-R intervals; therefore distinguishing preexcited AF from VT or preexcited SVT can be difficult. However, careful measurement always discloses definite irregularities. Moreover, very rapid and irregular VTs are usually unstable and quickly degenerate into VF. Thus, when a rapid, irregular wide QRS complex tachycardia is noted in a patient who has a reasonably stable hemodynamic state, preexcited AF is the most likely diagnosis.


The ability to conduct rapidly over an AV BT is determined primarily by the intrinsic conduction and refractoriness properties of the AV BT. However, as with AVN conduction, factors such as spatial and temporal characteristics of atrial wavefronts during AF, autonomic tone, and concealed conduction influence activation over the AV BT. Very rapid AV conduction during AF can occur in the presence of AV BTs with very short refractoriness, especially when normal conduction through the AVN and HPS is blocked (as occurs with AVN blocking drugs) and ventricular activation occurs only via the rapidly conducting BT, which would then eliminate retrograde concealment into the BT. This would result in extremely rapid ventricular rates, possibly more than 300 beats/min, which can occasionally degenerate into VF.


Regular Ventricular Rate During Atrial Fibrillation


Regular ventricular rate during AF indicates associated abnormalities. A regular, slow ventricular rhythm during AF suggests a junctional or ventricular rhythm, either as an escape mechanism with complete AV block or as an accelerated pacemaker activity with AV dissociation ( see Fig. 9.27 ). Rarely, the R-R interval can be regularly irregular and show group beating with the combination of complete heart block and a lower nodal pacemaker with a Wenckebach type of exit block. Patients with severe underlying heart disease may develop the combination of AF and VT, leading to a rapid, regular, wide QRS complex tachycardia.


Effect of Digitalis Toxicity on the Ventricular Response


With increasing degrees of digitalis toxicity, high-grade but not complete AV block during AF initially leads to single junctional or ventricular escape beats. Higher degrees of AV block result in such a small number of atrial impulses being conducted that the lower pacemaker takes over, thus leading to an escape junctional or ventricular rhythm with a regular R-R interval for two or more cycles. On occasion, the junctional rate can increase, possibly because of digitalis-induced triggered activity, and it is called nonparoxysmal junctional tachycardia. Increasing digitalis toxicity can result in a Wenckebach exit block and give the appearance of an irregular ventricular rhythm with restoration of AF conduction, but the rhythm shows repetitive group beating because of the exit block. Complete AV block is marked by a regular escape rhythm with no conducted beats, a finding that may lead to the erroneous assumption that the patient has converted to NSR. Infrequently, impulses from the lower pacemaker travel alternately down the right and left bundle branches or alternate fascicles of the left bundle branch, resulting in a bidirectional tachycardia. This arrhythmia, which is also frequently a reflection of marked digitalis toxicity, may appear to be ventricular bigeminy. In true bigeminy, however, the ventricular beat in the bigeminal pattern is premature. In comparison, the R-R interval is regular with a bidirectional tachycardia because all the beats arise from a single pacemaker.


QRS Morphology


The QRS complexes during AF are narrow and normal unless AV conduction is abnormal because of functional (rate-related) aberration, preexisting bundle branch block (BBB) (see Fig. 15.12 ), or preexcitation over an AV BT (see Fig. 15.14 ).


Aberrant conduction commonly occurs during AF. Aberrancy is caused by the physiological changes of the conduction system refractory periods that are associated with sudden changes in heart rate. The refractoriness of the HPS tissue is directly related to the preceding R-R interval. Thus there can be aberrant conduction from a long R-R interval followed by a short cycle. In this scenario, the refractory period of the bundles increases during the long R-R interval (long cycle). The QRS complex that ends the long pause will be conducted normally but is followed by a prolonged refractory period of the bundle branches. If the next QRS complex occurs after a short coupling interval, it can be conducted aberrantly because one of the bundle branches is still refractory due to a lengthening of the refractory period (the Ashman phenomenon). The gross irregularity of the ventricular response during AF yields an abundance of different R-R intervals; therefore the long-short cycle sequence occurs commonly, and the Ashman phenomenon is seen frequently during AF; right bundle branch block (RBBB) aberrancy is more common than left bundle branch block (LBBB) aberrancy, because the right bundle branch has a longer refractory period at slower heart rates. The left anterior fascicle is also frequently involved, often in combination with RBBB. In contrast, functional aberration is uncommon in the HB, the left posterior fascicle, or the main left bundle. Moreover, CLs preceding the pause may also affect the chance for aberrancy after the pause.


The aberrancy caused by the Ashman phenomenon can be present for one beat and have a morphology that resembles a PVC, or it can involve several sequential complexes, suggesting VT. The persistence of aberrancy may reflect a time-dependent adjustment of refractoriness of the bundle branch to an abrupt change in CL, or it can potentially be the result of concealed transseptal activation.


Although functional BBB is common in AF, PVCs are even more frequent, and it is important to differentiate between aberrant ventricular conduction and VT when repetitive wide QRS complexes occur during AF. The presence or absence of a long-short cycle sequence may not be helpful in differentiating aberration from ectopy for two reasons. Although a long cycle (pause) sets the stage for the Ashman phenomenon, it also tends to precipitate ventricular ectopy. Moreover, concealed conduction occurs frequently during AF, and therefore it is never possible to determine exactly when a bundle branch is activated from the surface ECG.


The proper diagnosis of aberrant conduction is a continuing challenge, but it can usually be accomplished by careful analysis of the rhythm strip and application of certain criteria. An aberrantly conducted beat caused by functional BBB generally has the pattern of a classic bundle branch or fascicular block. A PVC is usually followed by a longer R-R cycle, indicating the occurrence of a compensatory pause, the result of retrograde conduction into the AVN and anterograde block of the impulse originating in the atrium. The presence of long R-R cycles after the wide QRS complex that have identical CLs also suggests a ventricular origin ( see eFig. 10.2 ). Furthermore, the absence of a long-short cycle sequence associated with the wide or aberrant QRS complex suggests that it is of ventricular origin. Aberrancy is likely not present if, with inspection of a long ECG rhythm strip, there are R-R interval combinations that are longer and shorter than those associated with the wide QRS complex. Also, a ventricular origin is likely if there is a fixed coupling interval between the normal and wide QRS complexes ( Fig. 15.15 ).


Feb 22, 2019 | Posted by in CARDIOLOGY | Comments Off on Atrial Fibrillation

Full access? Get Clinical Tree

Get Clinical Tree app for offline access