Sei sulla pagina 1di 24

Atrial Fibrillation: Clinical Features,

Mechanisms, and Management


38
Fred Morady and Douglas P. Zipes

ELECTROCARDIOGRAPHIC FEATURES, 798 Aspirin and Other Antithrombotic NONPHARMACOLOGIC MANAGEMENT


Agents, 802 OF ATRIAL FIBRILLATION, 808
CLASSIFICATION OF ATRIAL
Warfarin, 803 Pacing to Prevent Atrial Fibrillation, 808
FIBRILLATION, 798
Newer Oral Anticoagulants, 803 Catheter Ablation of Atrial Fibrillation, 808
EPIDEMIOLOGY OF ATRIAL Low-Molecular-Weight Heparin, 804 Ablation of the Atrioventricular Node, 810
FIBRILLATION, 800 Excision or Closure of the Left Atrial Surgical Approaches to Atrial Fibrillation, 811
Appendage, 805
MECHANISMS OF ATRIAL SPECIFIC CLINICAL SYNDROMES, 811
FIBRILLATION, 800 ACUTE MANAGEMENT OF ATRIAL Postoperative Atrial Fibrillation, 811
FIBRILLATION, 805 Wolff-Parkinson-White Syndrome, 811
GENETIC FACTORS, 801
Heart Failure, 811
LONG-TERM MANAGEMENT OF ATRIAL
CAUSES OF ATRIAL FIBRILLATION, 801 Pregnancy, 812
FIBRILLATION, 806
CLINICAL FEATURES, 801 Pharmacologic Rate Control Versus Rhythm FUTURE PERSPECTIVES, 812
Control, 806
DIAGNOSTIC EVALUATION, 801 REFERENCES, 812
Pharmacologic Rate Control, 806
PREVENTION OF THROMBOEMBOLIC Pharmacologic Rhythm Control, 807 GUIDELINES, 813
COMPLICATIONS, 802 Rhythm Control with Agents Other Than
Risk Stratification, 802 Antiarrhythmic Drugs, 807

AF that is persistent for longer than 1 year is termed longstanding,


ELECTROCARDIOGRAPHIC FEATURES whereas longstanding AF refractory to cardioversion is termed per-
manent. However, “permanent AF” is not necessarily permanent in
Atrial fibrillation (AF) is a supraventricular arrhythmia characterized the literal sense because it may be eliminated successfully by surgical
electrocardiographically by low-amplitude baseline oscillations or catheter ablation.
(fibrillatory or f waves) and an irregularly irregular ventricular rhythm. Some patients with paroxysmal AF can occasionally have episodes
The f waves have a rate of 300 to 600 beats/min and are variable in that are persistent, and vice versa. The predominant form of AF
amplitude, shape, and timing. In contrast, flutter waves have a rate of determines how it should be categorized.
250 to 350 beats/min and are constant in timing and morphology A confounding factor in the classification of AF is cardioversion
(Fig. 38-1). In lead V1, f waves sometimes appear uniform and can and antiarrhythmic drug therapy. For example, if a patient undergoes
mimic flutter waves (Fig. 38-2). The distinguishing feature from atrial transthoracic cardioversion 24 hours after the onset of AF, it is
flutter is the absence of uniform and regular atrial activity in other unknown whether the AF would have persisted for more than 7 days.
leads of the electrocardiogram. In some patients, f waves are very Furthermore, antiarrhythmic drug therapy may change persistent AF
small and not perceptible on the electrocardiogram. In such patients into paroxysmal AF. It is generally thought that the classification of
the diagnosis of AF is based on the irregularly irregular ventricular AF should not be altered on the basis of the effects of electrical car-
rhythm (Fig. 38-3). dioversion or antiarrhythmic drug therapy.
The ventricular rate during AF in the absence of negative dromo- Lone AF refers to AF that occurs in patients younger than 60 years
tropic agents is typically 100 to 160 beats/min. In patients with Wolff- who do not have hypertension or any evidence of structural heart
Parkinson-White syndrome, the ventricular rate during AF can exceed disease. This designation is clinically relevant because patients with
250 beats/min because of conduction over the accessory pathway lone AF are at lower risk for thromboembolic complications, thus
(see Chapter 37). When the ventricular rate during AF is very rapid eliminating the necessity for anticoagulation with warfarin. In addi-
(>170 beats/min), the degree of irregularity is attenuated and the tion, the absence of structural heart disease allows the safe use of
rhythm can seem regular (Fig. 38-4). rhythm-control drugs such as flecainide in patients with lone AF.
The ventricular rhythm can be regular during AF in patients with Paroxysmal AF can also be classified clinically on the basis of the
a ventricular pacemaker who are fully paced and when a third-degree autonomic setting in which it most often occurs. Approximately 25%
atrioventricular (AV) block with a regular escape rhythm is present of patients with paroxysmal AF have vagotonic AF, in which AF is
(Fig. 38-5). In these cases the diagnosis of AF is based on the pres- initiated in the setting of high vagal tone, typically in the evening
ence of f waves. When there is a third-degree AV block with a junc- when the patient is relaxing or during sleep. Drugs that have a vago-
tional escape, a Wenckebach exit block in the AV junction (as can tonic effect (such as digitalis) can aggravate vagotonic AF, and drugs
occur during digitalis toxicity) results in a regularly irregular ventricu- that have a vagolytic effect (such as disopyramide) may be particu-
lar rate (see Chapters 34 and 37). larly appropriate for prophylactic therapy. Adrenergic AF occurs in
approximately 10% to 15% of patients with paroxysmal AF in the
setting of high sympathetic tone, for example, during strenuous exer-
CLASSIFICATION OF ATRIAL FIBRILLATION tion. In patients with adrenergic AF, beta blockers not only provide
rate control but can also prevent the onset of AF. Most patients have
AF that terminates spontaneously within 7 days is termed paroxysmal, a mixed or random form of paroxysmal AF, with no consistent pattern
and AF present continuously for more than 7 days is called persistent. of onset.

798 Additional content is available online at ExpertConsult.


799

38
V1

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


II

V5

V1

II

V5

FIGURE 38-1 Comparison of the f waves of AF (top panel) and the flutter waves of atrial flutter (bottom panel). Note that f waves are variable in rate, shape, and amplitude
whereas flutter waves are constant in rate and all aspects of morphology. Shown are leads V1, II, and V5.

V1

II

V5

FIGURE 38-2 Example of AF with prominent f waves in V1 that mimic atrial flutter waves. Note that typical f waves are present in leads II and V5, thereby establishing the
diagnosis of AF.

I aVR V1 V4

II aVL V2 V5

III aVF V3 V6

FIGURE 38-3 A 12-lead electrocardiogram of AF in which f waves are not discernible. The irregularly irregular ventricular rate indicates that this is AF and not a
junctional rhythm.
800

V V1
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

II

V5

FIGURE 38-4 Recording of AF with a rapid ventricular rate of 160 beats/min. Shown are leads V1, II, and V5. On quick review there may appear to be a regular rate consistent
with paroxysmal supraventricular tachycardia. On closer inspection, however, it is clear that the rate is irregularly irregular.

I aVR V1 V4

II aVL V2 V5

III aVF V3 V6

VI

II

V5

FIGURE 38-5 Atrial fibrillation with complete heart block and a regular junctional rhythm at a rate of 45 beats/min.

EPIDEMIOLOGY OF ATRIAL FIBRILLATION increased significantly between 1980 and 2000 from 4.4 to 5.4 in men
and from 2.4 to 2.8 in women.4 There was a relative increase of 0.6% per
AF is the most common arrhythmia treated in clinical practice and year in the age-adjusted incidence of AF. An increase in obesity
the most common arrhythmia for which patients are hospitalized; accounted for 60% of the age-adjusted increase in AF incidence. The
approximately 33% of arrhythmia-related hospitalizations are for AF. number of patients with AF in the United States was estimated to be 3.2
AF is associated with an approximately fivefold increase in the risk million in 1980 and 5.1 million in 2000 and was projected to be 12.1 to
for stroke and a twofold increase in the risk for all-cause mortality.1 15.9 million in 2050, all of which are higher than previous estimates.
AF is also associated with the development of heart failure.
Estimates of the actual number of individuals with AF in the United
States range between 2.3 and 5 million in most studies. The incidence MECHANISMS OF ATRIAL FIBRILLATION
of AF is age and sex related and ranges from 0.1% per year before
the age of 40 years to higher than 1.5% per year in women and The mechanisms responsible for AF are complex. Triggering events
higher than 2% per year in men older than 80 years. Heart failure, may differ from maintenance mechanisms. In addition, the clinical
aortic and mitral valve disease, left atrial enlargement, hypertension, phenotypes of paroxysmal, persistent, and longstanding persistent
and advanced age are independent risk factors for the development AF have different electrophysiologic characteristics because of
of AF, as are obesity and obstructive sleep apnea2 (see Chapter 75). remodeling and different clinical modulators that affect the substrate,
Another risk factor is psoriasis, which when severe, triples the risk for such as heart failure, atrial stretch and ischemia, sympathovagal
AF in patients younger than 50 years.3 influences, inflammation, and fibrosis.
A community-based cohort study in Olmstead County, Minnesota, There are probably two electrophysiologic mechanisms of AF:
reported that the age-adjusted incidence of AF per 1000 person-years one or more automatic, triggered, or microreentrant foci, so-called
801
drivers, which fire at rapid rates and cause fibrillation-like activity, CLINICAL FEATURES
and multiple reentrant circuits meandering throughout the atria 38
that annihilate and reform wavelets, thereby perpetuating the fibril- The symptoms of AF vary widely between patients and range from none

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


lation. In many studies the left atrium contains the site of dominant to severe and functionally disabling. The most common symptoms of
frequency discharge, with a left-to-right gradient. Both mechanisms AF are palpitations, fatigue, dyspnea, effort intolerance, and lighthead-
may be present simultaneously. In a recent study, computational edness. Polyuria can occur because of the release of atrial natriuretic
maps were obtained in patients by signal processing of multiple elec- hormone. Many patients with symptomatic paroxysmal AF also have
trograms recorded simultaneously during AF.5 This technique can asymptomatic episodes, and some patients with persistent AF have
reveal electrical rotors and focal sources. A mean of 2.1 sources was symptoms only intermittently, thus making it difficult to accurately
found in 97% of 101 patients, with 70% being rotors and 30% being assess the frequency and duration of AF on the basis of symptoms.
focal sources. It is estimated that approximately 25% of patients with AF are
Rapid discharges from the pulmonary veins are the most common asymptomatic, more commonly elderly patients and those with per-
triggers of AF and may also play a perpetuating role, more so in sistent AF. Such patients are sometimes erroneously classified as
paroxysmal AF than in persistent AF. This is why pulmonary vein having asymptomatic AF despite the presence of fatigue or effort
isolation is particularly effective for elimination of paroxysmal AF. In intolerance. Because fatigue is a nonspecific symptom, it may not be
persistent AF, changes in the atrial substrate, including interstitial clearly due to persistent AF. “Diagnostic cardioversion” may be
fibrosis, which contributes to slow, discontinuous, and anisotropic helpful by maintaining sinus rhythm for at least a few days to deter-
conduction, may give rise to complex fractionated atrial electrograms mine whether a patient feels better in sinus rhythm. This can provide
(CFAEs) and reentry. Therefore, pulmonary vein isolation is rarely a basis to pursue a rhythm-control versus rate-control strategy.
sufficient to eliminate persistent AF, and additional ablation of the Syncope is an uncommon symptom of AF. It can be caused by a
atrial substrate is usually necessary. long sinus pause on termination of AF in a patient with sick sinus
syndrome. Syncope can also occur during AF with a rapid ventricular
rate either because of neurocardiogenic (vasodepressor) syncope
GENETIC FACTORS that is triggered by the tachycardia or because of a severe drop in
blood pressure secondary to a reduction in cardiac output.
Several mutations that are responsible for familial AF and that predis- Asymptomatic or minimally symptomatic AF patients are not
pose to AF have been identified.6 These mutations cause a gain of prompted to seek medical care and can initially be seen with a
function of repolarization potassium currents that results in shorten- thromboembolic complication such as stroke or the insidious onset
ing of atrial refractoriness and facilitation of atrial reentry. Multiple of heart failure symptoms that eventually results in florid congestive
polymorphisms that are associated with AF that is idiopathic, are heart failure.
associated with structural heart disease, or occur postoperatively The hallmark of AF on physical examination is an irregularly irreg-
have also been identified.6 These polymorphisms are in genes that ular pulse. Short R-R intervals during AF do not allow adequate time
affect potassium and sodium channels, sarcolipin, the renin- for left ventricular diastolic filling, which results in a low stroke
angiotensin system, connexin 40, endothelial nitric oxide synthase, volume and the absence of palpable peripheral pulse. This leads to
and interleukin-10. The end results are changes in calcium handling, a “pulse deficit,” during which the peripheral pulse is not as rapid as
fibrosis, conduction, and inflammation that predispose to AF. the apical rate. Other manifestations of AF on physical examination
are irregular jugular venous pulsations and variable intensity of the
first heart sound.
CAUSES OF ATRIAL FIBRILLATION
Most patients with AF have hypertension (usually with left ventricular DIAGNOSTIC EVALUATION
hypertrophy; see Chapters 43 and 44) or some other form of struc-
tural heart disease. In addition to hypertensive heart disease, the In a patient who describes irregular or rapid palpitations suggestive
most common cardiac abnormalities associated with AF are isch- of paroxysmal AF, ambulatory monitoring is useful to document
emic heart disease, mitral valve disease, hypertrophic cardiomyopa- whether AF is responsible for the symptoms. If the symptoms occur
thy, and dilated cardiomyopathy. Less common causes of AF are on a daily basis, a 24-hour Holter recording is appropriate. However,
restrictive cardiomyopathies such as amyloidosis, constrictive peri- extended monitoring for 2 to 4 weeks with an event monitor or by
carditis, and cardiac tumors. Severe pulmonary hypertension is often mobile cardiac outpatient telemetry is appropriate for patients whose
associated with AF. symptoms are sporadic (see Chapter 34).
Obesity and obstructive sleep apnea (see Chapter 75) are associ- The history should be directed at determination of the type and
ated with each other, and both have been found to independently severity of symptoms, the first onset of AF, whether the AF is parox-
increase the risk for AF.2 The data available suggest that atrial dilation ysmal or persistent, triggers for AF, whether the episodes are random
and an increase in systemic inflammatory factors are responsible for or occur at particular times (such as during sleep), and the frequency
the relationship between obesity and AF. Possible mechanisms of AF and duration of episodes. When it is unclear from the history, 2 to 4
in patients with sleep apnea include hypoxia, surges in autonomic weeks of ambulatory monitoring with an autotrigger event monitor
tone, and hypertension. or by mobile cardiac outpatient telemetry is useful to determine
AF can be due to causes that are temporary or reversible. The most whether the AF is paroxysmal or persistent and to quantitate the AF
common temporary causes are excessive alcohol intake (holiday burden in patients with paroxysmal AF. The history also should be
heart), open heart or thoracic surgery, myocardial infarction, pericar- directed at identification of potentially correctible causes (e.g., hyper-
ditis (see Chapter 71), myocarditis, and pulmonary embolism (see thyroidism, excessive alcohol intake), structural heart disease, and
Chapter 73). The most common correctable cause is hyperthyroid- comorbid conditions.
ism (see Chapter 81). Laboratory testing should include thyroid function tests, liver func-
AF is sometimes induced by tachycardia. Patients with tachycardia- tion tests, and renal function tests. Echocardiography is always
induced AF most often have AV nodal reentrant tachycardia or a appropriate to evaluate atrial size and left ventricular function and
tachycardia related to Wolff-Parkinson-White syndrome that degener- to look for left ventricular hypertrophy, congenital heart disease
ates into AF. If a patient with AF has a history of rapid and regular (see Chapter 62), and valvular heart disease. Chest radiography is
palpitations before the onset of irregular palpitations or has a Wolff- appropriate if the history or findings on physical examination are
Parkinson-White electrocardiographic pattern, this should raise sus- suggestive of pulmonary disease (see Chapter 15). A stress test is
picion for tachycardia-induced AF. Treatment of the tachycardia that appropriate for evaluation of ischemic heart disease in at-risk patients
triggers the AF often but not always prevents recurrences of AF. (see Chapter 13).
802
of heart failure and advanced age. Therefore, it may be appropriate
PREVENTION OF THROMBOEMBOLIC to take renal failure into account in evaluating the risk profile of a
V
COMPLICATIONS patient with AF.
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

By definition, the burden of AF is greater in patients with persistent


Risk Stratification AF than in those with paroxysmal AF. It may seem reasonable to
A major goal of therapy in patients with AF is to prevent thromboem- assume that the risk for stroke is lower in patients with occasional
bolic complications such as stroke. It is well established that warfarin episodes of self-limited AF than in those with AF continuously.
is more effective than aspirin for prevention of thromboembolic com- However, the data available in fact indicate that the risk for thrombo-
plications.7 However, because of the risk for hemorrhage during war- embolic complications is the same in patients with paroxysmal and
farin therapy, its use should be limited to patients whose risk for persistent AF. Even 15 minutes of AF may be long enough to result in
thromboembolic complications is greater than their risk for hemor- local cardiac platelet activation and endothelial dysfunction, which
rhage. Therefore, it is useful to risk-stratify patients with AF to identify predispose to thrombus formation during an acute episode of AF.12
appropriate candidates for warfarin therapy. Therefore, the type of AF should not be taken into account in risk-
The strongest predictors of ischemic stroke and systemic thrombo- stratifying AF patients for thromboembolic risk.
embolism are a history of stroke or a transient ischemic episode and Modern-day dual-chamber pacemakers and implantable
mitral stenosis. When patients with AF and a previous ischemic cardioverter-defibrillators (ICDs) are capable of detecting short epi-
stroke are treated with aspirin, the risk for another stroke is very high, sodes of asymptomatic AF that would otherwise not have been
in the range of 10% to 12% per year. At the other end of the risk spec- detected clinically. In a recent multicenter prospective study, sub-
trum are patients with lone AF, whose cumulative 15-year risk for clinical atrial tachyarrhythmias (atrial rate >190 beats/min for >6
stroke was reported to be in the range of 1% to 2%. Aside from previ- minutes) were detected by device interrogation in 10.1% of patients
ous stroke, the best-established risk factors for stroke in patients with 65 years or older with hypertension and no history of AF who received
nonvalvular AF are diabetes (relative risk, 1.7), hypertension (relative a pacemaker or ICD.13 Subclinical atrial tachyarrhythmias were inde-
risk, 1.6), heart failure (relative risk, 1.4), and age 70 years or older pendently associated with a 2.5-fold increase in the risk for stroke.
(relative risk, 1.4 per decade).8 An important consideration in patients treated with an oral antico-
A simple clinical scheme to risk-stratify patients on the basis of agulant is the risk for bleeding. Several risk stratification scoring
major risk factors is the CHADS2 (cardiac failure, hypertension, age, systems have been developed to assess a patient’s susceptibility to
diabetes, stroke) score. Each of the first four risk factors is worth 1 hemorrhagic complications. The scoring system with the best balance
point, and a previous stroke or transient ischemic event is worth 2 of simplicity and accuracy is the HAS-BLED score.14 The components
points. There is a direct relationship between the CHADS2 score and of this score are hypertension, abnormal renal or liver function,
the annual risk for stroke in the absence of aspirin or warfarin therapy. stroke, bleeding history or predisposition, labile international normal-
The clinical value of the CHADS2 score lies in its simplicity and pre- ized ratio (INR), older age (>75 years), and concomitant drug (anti-
dictive value. However, recent studies have demonstrated that the platelet agent or nonsteroidal anti-inflammatory drug) or alcohol use.
CHA2DS2-VASc score more accurately discriminates low-risk from Each of these components is worth 1 point. As the score increases
intermediate-risk patients.9 from 0 to the maximum of 9, there is a stepwise increase in the risk
In this risk score system, cardiac failure, hypertension, diabetes, for bleeding in patients treated with warfarin. For example, in one
vascular disease, 65 to 74 years of age, and female sex are worth 1 study the annual rate of major bleeding was 1.1% in patients with a
point each, whereas age 75 years or older and previous stroke or HAS-BLED score of 0, 3.7% with a score of 3, and 12.5% with a score
transient ischemic event are worth 2 points. The annual risk for stroke of 5.15
is zero or close to zero when the CHA 2DS2-VASc score is 0, as opposed In two large-scale cohort studies totaling 132,372 patients16 and
to approximately 2% when the CHADS2 score is 0.10 A score of 1 is 170,292 patients17 with nonvalvular AF, the CHA 2DS2-VASc and HAS-
associated with an annual stroke risk of approximately 3% with the BLED scores were calculated for each patient. The net clinical benefit
CHADS2 score versus 0.7% with the CHA 2DS2-VASc score (Fig. 38-6). of warfarin was defined as the number of strokes while not taking
A large-scale study demonstrated that renal failure is also an inde- warfarin minus the number of intracranial bleeding episodes while
pendent risk factor for stroke in patients with AF.11 The relative risk taking warfarin. In both studies, warfarin was associated with a net
for a thromboembolic event in the absence of anticoagulation was clinical benefit except when the CHA2DS2-VASc score was 0. In
1.4 in patients with an estimated glomerular filtration rate lower than patients with a CHA 2DS2-VASc score of 1 or higher, the risk for stroke
45 mL/min/1.73 m2. The predictive strength of this degree of renal in the absence of warfarin exceeded the number of bleeding compli-
failure for a thromboembolic event appears to be equivalent to that cations during treatment with warfarin.
The results of these large cohort studies notwithstanding, the deci-
sion to institute anticoagulation in a patient in clinical practice should
20 be individualized. At times it may be appropriate to not initiate anti-
CHADS2 coagulation in a patient with a CHA 2DS2-VASc score of 1 or higher.
18
CHA2DS2-VASc For example, the annual risk for stroke in a patient with a CHA 2DS2-
ANNUAL STROKE RATE

16 VASc score of 2 is approximately 2%, which usually justifies the use


14 of warfarin. However, if that patient has a HAS-BLED score of 5 or
higher, which is associated with an annual risk for major bleeding of
12 12% or higher, it would be imprudent to treat that patient with
10 warfarin.
It should be noted that the HAS-BLED score was developed and
8
validated in patients in whom warfarin was used for anticoagulation.
6 Except for labile INR, it is likely that the components of the HAS-BLED
4 score also apply to patients in whom a direct thrombin inhibitor or
factor Xa inhibitor is used for anticoagulation. However, the predic-
2 tive value of the HAS-BLED score in patients treated with one of the
0 newer antithrombotic agents has not yet been determined.
0 1 2 3 4 5 6 7 8 9
FIGURE 38-6 Annual risk for stroke (percent risk per year) based on the CHADS2 Aspirin and Other Antithrombotic Agents
and CHA2DS2-VASc scores. (Based on data from Lip GY: Implications of the CHA[2]
DS[2]-VASc and HAS-BLED scores for thromboprophylaxis in atrial fibrillation. Am J Aspirin does not prevent thromboembolic complications as effec-
Med 124:111, 2011.) tively as warfarin does in patients with AF. In a meta-analysis of five
803
randomized clinical trials, aspirin reduced the risk for stroke by only with warfarin. The fear of hemorrhagic complications may lead
18%.7 In a recent large cohort study of patients with nonvalvular AF, some clinicians to favor the use of aspirin over warfarin in older 38
aspirin had no therapeutic efficacy in preventing strokes.16 Therefore, adults. However, recent data indicate that the risk-to-benefit ratio of

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


if aspirin is used for prophylactic therapy, it should be used only in warfarin is more favorable than that of aspirin, even in patients older
patients at lowest risk for thromboembolic complications (CHA 2DS2- than 75 years. A randomized clinical trial (the Birmingham Atrial
VASc score of 0). The 2011 American College of Cardiology/American Fibrillation Treatment of the Aged Study) enrolled 973 patients older
Heart Association/Heart Rhythm Society guidelines still recommend than 75 years (mean age, 82 years) with AF and randomly assigned
aspirin for stroke prevention in patients with a CHADS2 score of 0 and them to treatment with 75 mg/day of aspirin or with warfarin adjusted
either aspirin or an oral anticoagulant when the CHADS2 score is 1.18 to maintain an INR of 2.0 to 3.0.22 The primary endpoint was the
Because of the negligible therapeutic effect of aspirin, a risk for bleed- composite of stroke (ischemic or hemorrhagic), intracranial hemor-
ing complications that is close to the risk associated with oral anti- rhage, and arterial embolism, and the mean duration of follow-up
coagulants, and the ability of the CHA 2DS2-VASc score to accurately was 2.7 years. The annual risk for the composite endpoint was signifi-
identify low-risk patients, the most recent guidelines of the European cantly higher in the aspirin group (3.8%) than in the warfarin group
Society of Cardiology recommend no antithrombotic therapy when (1.8%), even when the analysis was limited to patients older than 85
the CHA 2DS2-VASc score is 0 and an individualized decision regard- years. These data suggest that age should not be considered a con-
ing no antithrombotic therapy versus an oral anticoagulant when the traindication to treatment with warfarin in patients with AF.
CHA 2DS2-VASc score is 1.19 It is well established that genetic factors influence the dose of
If aspirin is used for stroke prevention in patients with AF, the warfarin required to maintain the INR within the therapeutic range.
appropriate daily dose is 81 to 325 mg/day. No data are available Several single nucleotide polymorphisms that affect warfarin metab-
to indicate superiority of a particular dose for prevention of olism have been identified. Algorithms based on pharmacogenetic
thromboembolism. (see Chapter 9) and clinical factors improve the accuracy of warfa-
In patients with a CHADS2 score higher than 1 who are not able to rin dose initiation more than do algorithms based only on clinical
tolerate anticoagulation with warfarin, combination therapy with factors, particularly for outliers who require 21 mg/wk or less or
aspirin and the platelet inhibitor clopidogrel is more efficacious than 49 mg/wk or more of warfarin to maintain a therapeutic INR.23
aspirin alone for prevention of thromboembolic complications. In a However, a randomized study demonstrated that warfarin dosing that
randomized, double-blind clinical trial (ACTIVE-A), all patients with took into account the results of genotyping for CYP2C9 (a cytochrome
AF and one or more risk factors for stroke who were not suitable P-450 isoform) and VKORC1 (a vitamin K epoxide reductase complex
candidates for anticoagulation with warfarin were treated with 75 to subunit) did not improve the time in the therapeutic range, which
100 mg/day of aspirin.20 The patients were randomly assigned to also was approximately 70% in both groups.24 Additional studies are
receive either 75 mg/day of clopidogrel or a matching placebo. The required to determine whether the clinical benefits of genotyping of
primary outcome was a composite of stroke, myocardial infarction, warfarin candidates justify the cost of genetic testing.
systemic embolism, and vascular death. When compared with
placebo, clopidogrel reduced the risk for stroke by 28% and risk for
the primary outcome by 11% but increased the risk for major hemor- Newer Oral Anticoagulants
rhage. The study demonstrated that for every 1000 patients treated Direct thrombin inhibitors and factor Xa inhibitors have several
with the combination of aspirin plus clopidogrel instead of aspirin advantages over vitamin K antagonists such as warfarin, the most
alone, 28 strokes (17 fatal or disabling) and 6 myocardial infarctions notable being a fixed dosing regimen, which eliminates the need for
would be prevented, at a cost of 20 major bleeding episodes (3 fatal). monitoring of a laboratory test such as the INR. Dabigatran, an oral
Therefore, in high-risk patients who are not suitable candidates for direct thrombin inhibitor, and rivaroxaban, a factor Xa inhibitor, were
warfarin, the benefits of combination therapy with aspirin plus clopi- approved by the Food and Drug Administration for prevention of
dogrel outweigh the risk. stroke/embolism in patients with nonvalvular AF in 2010 and 2011,
respectively. Another factor Xa inhibitor, apixaban, was expected to
gain Food and Drug Administration approval in 2013.
Warfarin Randomized clinical trials have demonstrated that each of these
A meta-analysis of the major randomized clinical trials in which three new oral anticoagulants is noninferior or superior to warfarin
warfarin was compared with placebo for prevention of thromboem- in efficacy and safety. These studies included patients with nonval-
bolism in patients with AF demonstrated that warfarin reduced the vular AF who had risk factors for stroke. In the RE-LY study, dabiga-
risk for all strokes (ischemic and hemorrhagic) by 61%.7 The target tran at a dose of 150 mg twice daily was associated with a lower risk
INR should be 2.0 to 3.0. This range of INRs provides the best balance for stroke and systemic embolism than warfarin was and a similar
between stroke prevention and hemorrhagic complications. In clini- rate of major hemorrhage.25 In the ROCKET-AF study, rivaroxaban at
cal practice, maintenance of the INR in the therapeutic range has a dose of 20 mg once daily was noninferior to warfarin for prevention
been challenging, and a large proportion of patients often have an of stroke/systemic embolism and was associated with a risk for major
INR lower than 2.0. A large prospective study of community-based bleeding that did not differ from that of warfarin.26 However, intracra-
practices demonstrated that the mean time in the therapeutic range nial hemorrhage and fatal bleeding were less common with rivaroxa-
in patients treated with warfarin was just 66% and that the time in the ban. In the ARISTOTLE study, apixaban at a dose of 5 mg twice daily
therapeutic range was less than 60% in 34% of patients.21 Maintaining was superior to warfarin in prevention of stroke/systemic embolism
the INR at a level of 2.0 or higher is important because even a rela- and was associated with a lower risk for hemorrhagic complications
tively small decrease in the INR from 2.0 to 1.7 more than doubles the and lower mortality (Fig. 38-7).27
risk for stroke. Furthermore, the data available indicate that the com- The newer oral anticoagulants, in addition to eliminating the need
bination of aspirin and low-intensity anticoagulation with warfarin is for laboratory monitoring, have other advantages over warfarin: fewer
inferior to warfarin in the standard therapeutic range for stroke drug interactions, no food interactions, and a rapid onset of action
prevention. that obviates the need for bridging therapy. However, they also have
The annual risk for a major hemorrhagic complication during anti- some disadvantages in comparison to warfarin: higher cost, more
coagulation with warfarin is in the range of 1% to 2%, and a strong gastrointestinal side effects in the case of dabigatran, twice-daily
predictor of major bleeding events is an INR higher than 3.0. For dosing in the case of dabigatran and apixaban, and absence of a
example, the risk for intracranial bleeding is approximately twice as laboratory test to verify compliance. Furthermore, these agents
high at an INR of 4.0 than at 3.0. This emphasizes the importance of cannot be used safely in patients with severe renal disease. Another
maintaining the INR in the range of 2.0 to 3.0. limitation is that the effects of the newer anticoagulants may be dif-
Some studies have indicated that advanced age can be a risk ficult to reverse in patients with an overdose or hemorrhage. For
factor for intracranial hemorrhage in patients with AF who are treated example, in a 2011 study, a single bolus of 50 IU/kg of prothrombin
804
Total mortality used in patients with a creatinine
V clearance lower than 30 mL/min.
NOACs VKAs Risk Ratio Risk Ratio
Study or Subgroup Events Total Events Total Weight M-H, Fixed, 95% Cl M-H, Fixed, 95% Cl
An issue that has not been
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

addressed in a randomized clini-


NCT01136408(D) 0 104 0 62 Not estimable cal trial is whether the newer oral
PETRO 0 166 0 70 Not estimable anticoagulants provide adequate
RE-LY 884 12091 487 6022 41.1% 0.90 (0.81, 1.01)
protection from the thromboem-
WEITZ 11 713 3 250 0.3% 1.29 (0.36, 4.57)
CHUNG 1 159 1 75 0.1% 0.47 (0.03, 7.44) bolic complications of transtho-
YAMASHITA 1 260 1 125 0.1% 0.48 (0.03, 7.62) racic cardioversion. Although
ARISTOTLE-J 0 148 0 74 Not estimable not studied prospectively, the
ARISTOTLE 603 9120 669 9081 42.4% 0.90 (0.81, 1.00) safety of dabigatran in patients
NCT00973245 (R1) 0 75 0 27 Not estimable undergoing cardioversion was
NCT00973323 (R2) 0 50 0 26 Not estimable evaluated in a post hoc analysis
J-ROCKET-AF 7 637 5 637 0.3% 1.40 (0.45, 4.39) of the RE-LY study.30 A subset
ROCKET-AF 208 7061 260 7082 15.8% 0.83 (0.70, 1.00) of 1336 patients underwent
cardioversion after 3 weeks or
Total (95%, Cl) 30584 23531 100.0% 0.89 (0.83, 0.96)
Total events 1715 1416 more of treatment with dabiga-
Heterogeneity: Chi2 = 1.91, df = 6 (P = 0.93); I2 = 0% 0.01 0.1 1 10 100 tran, 150 mg twice daily, or dose-
A Test for overall effect: Z = 3.24 (P = 0.001) Favors NOACs Favors VKAs adjusted warfarin with an INR of
2.0 to 3.0. The stroke/systemic
Cardiovascular mortality embolism rate at 30 days did
NOACs VKAs Risk Ratio Risk Ratio not differ significantly between
Study or Subgroup Events Total Events Total Weight M-H, Fixed, 95% Cl M-H, Fixed, 95% Cl the dabigatran group (0.3%)
and the warfarin group (0.6%).
NCT01136408 (D) 0 104 0 62 Not estimable
There was also no difference
PETRO 0 166 0 70 Not estimable
RE-LY 563 12091 317 6022 43.8% 0.88 (0.77, 1.01) between the two groups in the
WEITZ 6 713 2 250 0.3% 1.05 (0.21, 5.18) rate of major bleeding (0.6% in
CHUNG 1 159 0 75 0.1% 1.43 (0.06, 34.57) both groups). These data suggest
YAMASHITA 0 260 0 125 Not estimable that dabigatran is a safe and
ARISTOTLE-J 0 148 0 74 Not estimable effective alternative to warfarin
ARISTOTLE 308 9120 344 9081 35.7% 0.89 (0.77, 1.04) in patients requiring cardiover-
NCT00973245 (R1) 0 75 0 27 Not estimable sion. However, because patient
NCT00973323 (R1) 0 50 0 26 Not estimable compliance and a therapeutic
J-ROCKET-AF 6 637 2 637 0.2% 3.00 (0.61, 14.81)
effect of dabigatran cannot be
ROCKET-AF 170 7061 193 7082 19.9% 0.88 (0.72, 1.08)
confirmed by laboratory testing,
Total (95%, Cl) 30584 23531 100.0% 0.89 (0.82, 0.98) a precardioversion transesopha-
Total events 1054 858 geal echocardiogram to rule
Heterogeneity: Chi2 = 2.36, df = 5 (P = 0.80); I2 = 0% 0.01 0.1 1 10 100 out a left atrial thrombus may
B Test for overall effect: Z = 2.50 (P = 0.001) Favors NOACs Favors VKAs be appropriate more often in
patients treated with dabigatran
FIGURE 38-7 Total (A) and cardiovascular (B) mortality during oral anticoagulant treatment. ARISTOTLE = Apixaban for the
Prevention of Stroke in Subjects with Atrial Fibrillation; CI = confidence interval; J-ARISTOTLE = Japanese Apixaban for the Preven- than in those treated with
tion of Stroke in Subjects with Atrial Fibrillation; J-ROCKET-AF = An Efficacy and Safety Study of Rivaroxaban with Warfarin for warfarin.
the Prevention of Stroke and Non-Central Nervous System Systemic Embolism in Patients with Non-Valvular Atrial Fibrillation in The onset of action of dabiga-
Japan; M-H = Mantel-Haenszel; NCT = National Clinical Trials database; NOACs = novel oral anticoagulants; RE-LY = Randomized tran, rivaroxaban, and apixaban
Evaluation of Long-Term Anticoagulant Therapy; ROCKET-AF = An Efficacy and Safety Study of Rivaroxaban with Warfarin for the
Prevention of Stroke and Non-Central Nervous System Systemic Embolism in Patients with Non-Valvular Atrial Fibrillation; VKAs is approximately 1.5 to 2 hours
= vitamin K antagonists. (From Dentali F, Riva N, Crowther M, et al: Efficacy and safety of the novel oral anticoagulants in atrial after a dose. The half-lives of
fibrillation: A systematic review and meta-analysis of the literature. Circulation 126:2381, 2012.) dabigatran and apixaban range
between 10 and 16 hours, and
the half-life of rivaroxaban is 5 to
complex concentrate was demonstrated to quickly and completely 9 hours. These anticoagulants lose most of their effect by 24 hours
reverse the anticoagulant effect of rivaroxaban, but not dabigatran.28 after discontinuation. The rapid onset of action and washout elimi-
Nonetheless, for many patients with AF, the advantages of the newer nate the need for bridging therapy with heparin when treatment with
anticoagulants outweigh the disadvantages. one of the new anticoagulants is interrupted for a surgical or invasive
The major professional societies have incorporated recommenda- procedure. In a recent study of patients treated with dabigatran who
tions regarding the use of factor Xa and/or direct thrombin inhibitors underwent radiofrequency catheter ablation of AF, dabigatran was
into their most recent updates of guidelines for the management of withheld on the morning of the procedure.31 The study included a
AF. The practice guidelines of the American College of Cardiology/ comparison group of patients who underwent radiofrequency cath-
American Heart Association/Heart Rhythm Society recommend eter ablation of AF during uninterrupted therapy with warfarin at an
dabigatran as a useful alternative to warfarin for prevention of stroke/ INR of 2.0 to 3.0. Major hemorrhagic complications occurred signifi-
systemic embolism in patients with nonvalvular paroxysmal or per- cantly more often in the dabigatran group (6%) than in the warfarin
sistent AF and risk factors for stroke. However, this recommendation group (1%). The results of this study demonstrate that dabigatran
is limited to patients without a prosthetic valve, with creatinine clear- should be withheld for at least 24 hours before an invasive or surgical
ance lower than 15 mL/min, or with impaired clotting function as a procedure.
result of advanced liver disease.29 The European Society of Cardiol-
ogy guidelines recommend dabigatran, rivaroxaban, or apixaban for
patients with AF in whom maintenance of a therapeutic INR during Low-Molecular-Weight Heparin
treatment with warfarin is difficult and state that one of these newer Low-molecular-weight heparin has a longer half-life than unfraction-
anticoagulants should be considered instead of dose-adjusted warfa- ated heparin does and a predictable antithrombotic effect that is
rin for most patients with nonvalvular AF, based on their net clinical attained with a fixed dosage administered subcutaneously twice a
benefits.19 The guidelines also recommend that these agents not be day. Because low-molecular-weight heparin can be self-injected by
805
patients outside the hospital, it is a practical alternative to unfraction- symptoms, previous AF episodes, age, left atrial size, and current
ated heparin for initiation of anticoagulation with warfarin in patients antiarrhythmic drug therapy. For example, in an elderly patient 38
with AF. Bridging therapy with low-molecular heparin should be con- whose symptoms resolve once the ventricular rate is controlled and

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


tinued until the INR is 2.0 or higher. who has already had early recurrences of AF despite rhythm-control
Because of its high cost, low-molecular-weight heparin is rarely drug therapy, further attempts at cardioversion are not usually appro-
used in clinical practice as a substitute for long-term conventional priate. On the other hand, cardioversion is generally appropriate for
anticoagulation. Low-molecular-weight heparin is typically used as a patients with symptomatic AF who are seen with a first episode of AF
temporary bridge to therapeutic anticoagulation when therapy with or who have had long intervals of sinus rhythm between previous
warfarin is initiated or in high-risk patients for a few days before and episodes.
after a medical or dental procedure when anticoagulation with war- If cardioversion is decided on for a hemodynamically stable patient
farin has been suspended. with AF that does not appear to be self-limited, two management
decisions must be made: early versus delayed cardioversion and
pharmacologic versus electrical cardioversion.
Excision or Closure of the Left The advantages of early cardioversion are rapid relief of symptoms,
Atrial Appendage avoidance of the need for transesophageal echocardiography or
Approximately 90% of left atrial thrombi form in the appendage, therapeutic anticoagulation for 3 to 4 weeks before cardioversion if
and therefore successful excision or closure of the left atrial append- it is performed within 48 hours of the onset of AF, and possibly a
age should markedly reduce the risk for thromboembolic complica- lower risk for early recurrence of AF because of less atrial remodeling
tions in patients with AF. Surgical techniques consist of either excision (see Chapter 33). A reason to defer cardioversion is the unavail-
or closure by suturing or stapling. The efficacy of these techniques ability of transesophageal echocardiography in an unanticoagulated
is variable and probably dependent on both technique and the patient with AF of unclear duration or a duration longer than 48
operator. Postoperative transesophageal echocardiography demon- hours. Other reasons include a left atrial thrombus noted on trans-
strated that the appendage was closed successfully in only 40% of esophageal echocardiography (see Fig. 15-91), a suspicion (based on
137 patients.32 The rate of successful closure was higher when the previous AF episodes) that AF will convert spontaneously within a
appendage was excised (73%) than when it was closed by suturing few days, or a correctable cause of AF (e.g., hyperthyroidism).
(23%). Of note is that complete closure was never achieved by sta- When cardioversion is performed early in the course of an episode
pling of the appendage. Left atrial appendage thrombi were never of AF, either pharmacologic or electrical cardioversion is an option.
seen on transesophageal echocardiography after excision of the Pharmacologic cardioversion has the advantage of not requiring
appendage but were observed in 41% of patients who underwent general anesthesia or deep sedation. In addition, the probability of
closure of the appendage. Therefore, transesophageal echocardiog- an immediate recurrence of AF may be lower with pharmacologic
raphy should be performed after surgical closure of the left atrial cardioversion than with electrical cardioversion. However, pharma-
appendage to confirm successful closure before discontinuation of cologic cardioversion is associated with a risk for adverse drug
anticoagulation. effects and is not as effective as electrical cardioversion. Pharmaco-
Closure of the left atrial appendage can also be achieved percuta- logic cardioversion is very unlikely to be effective if the duration of
neously with an implanted device intended to seal the appendage. A AF is longer than 7 days.
randomized clinical trial (PROTECT AF) compared the efficacy of a Drugs that can be administered intravenously for cardioversion of
percutaneous closure device versus warfarin for prevention of throm- AF consist of ibutilide, procainamide, and amiodarone. For AF epi-
boembolic complications in 707 patients with AF and a CHADS2 score sodes shorter than 2 to 3 days in duration, the efficacy of these drugs
of 1 or higher.33 The device was found to be noninferior to warfarin is approximately 60% to 70% for ibutilide, 40% to 50% for amiodarone,
for the primary efficacy composite endpoint of stroke, systemic and 30% to 40% for procainamide. To minimize the risk for QT pro-
emboli, and cardiovascular death and superior to warfarin for hemor- longation and polymorphic ventricular tachycardia (torsades de
rhagic stroke (91% reduction). The complication rate was approxi- pointes; see Chapter 37), use of ibutilide should be limited to
mately four times higher in the device arm, with the most common patients with an ejection fraction higher than 35%. Acute pharmaco-
complication being pericardial effusion. The study demonstrated that logic cardioversion of AF can also be attempted with orally adminis-
percutaneous closure of the left atrial appendage is an effective tered drugs in patients without structural heart disease. The most
alternative to warfarin in patients with AF. Another recent study commonly used oral agents for acute conversion of AF are propafe-
showed that the risk for pericardial effusion declines significantly as none (300 to 600 mg) and flecainide (100 to 200 mg). It is prudent to
operator experience increases.34 Approval of the left atrial append- administer these drugs under surveillance the first time that they are
age closure device by the Food and Drug Administration awaits addi- used. If no adverse drug effects are observed, the patient may then
tional safety data. be an appropriate candidate for episodic, self-administered antiar-
It is likely that the left atrial appendage closure device will have its rhythmic drug therapy on an outpatient basis.
greatest utility in high-risk patients with AF who cannot tolerate or The efficacy of transthoracic cardioversion is approximately 95%.
refuse to take an oral anticoagulant. Biphasic waveform shocks convert AF more effectively than do
monophasic waveform shocks and allow the use of lower energy
shocks, which result in a lower risk for skin irritation. An appropriate
first-shock strength using a biphasic waveform is 150 to 200 J followed
ACUTE MANAGEMENT OF by higher output shocks if needed. If a 360-J biphasic shock is unsuc-
ATRIAL FIBRILLATION cessful, ibutilide should be infused before another shock is delivered
because it lowers the defibrillation energy requirement and improves
Patients who go to the emergency department because of AF gener- the success rate of transthoracic cardioversion.
ally have a rapid ventricular rate, and control of the ventricular rate Two types of failure of transthoracic cardioversion occur in patients
is most rapidly achieved with intravenous diltiazem or esmolol. If the with AF. The first type is complete failure to restore sinus rhythm. In
patient is hemodynamically unstable, immediate transthoracic car- this situation, an increase in shock strength or infusion of ibutilide
dioversion may be appropriate. If the AF has been present for more often results in successful cardioversion. The second type of failure
than 48 hours or if the duration is unclear and the patient is not is immediate recurrence of AF within a few seconds of successful
already receiving an anticoagulant, cardioversion should be pre- conversion to sinus rhythm. The incidence of immediate recurrence
ceded by transesophageal echocardiography to rule out a left atrial of AF is approximately 25% for episodes shorter than 24 hours in
thrombus. duration and approximately 10% for episodes longer than 24 hours
If the patient is hemodynamically stable, the decision to restore in duration. For this type of failure of cardioversion, an increase in
sinus rhythm by cardioversion is based on several factors, including shock strength is of no value. If the patient has not been receiving an
806
oral rhythm-control agent, infusion of ibutilide may be helpful for effects and efficacy of the antiarrhythmic drugs already used to treat
V prevention of an immediate recurrence of AF. the patient; and the patient’s preference.
Regardless of whether cardioversion is performed pharmacologi- The AFFIRM study convincingly demonstrated that a rate-control
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

cally or electrically, therapeutic anticoagulation is necessary for 3 strategy is preferable to a rhythm-control strategy in asymptomatic or
weeks or longer before cardioversion to prevent thromboembolic minimally symptomatic patients 65 years or older. In patients with
complications if the AF has been ongoing for more than 48 hours. If persistent AF, it is reasonable to attempt to restore sinus rhythm with
the time of onset of AF is unclear, for the sake of safety, the duration antiarrhythmic drug therapy or transthoracic cardioversion at least
of AF should be assumed to be greater than 48 hours. These patients once in individuals 65 years or younger and in those 65 years or older
should receive therapeutic anticoagulation for 4 weeks after cardio- whose AF is symptomatic despite adequate heart rate control. If the
version to prevent the thromboembolic complications that may occur AF has been continuous for longer than 1 year or if the left atrial
because of atrial stunning. If the duration of AF is known to be less diameter is very large (>5.0 cm), there is a high probability of an early
than 48 hours, cardioversion can be performed without anticoagula- recurrence of AF, and this should be taken into account in deciding
tion. To improve the safety margin, it may be appropriate to use a on the best strategy. After cardioversion, the decision to maintain the
24-hour cutoff for the AF duration, which allows safe cardioversion patient on antiarrhythmic drug therapy to delay the next episode of
without anticoagulation. AF is based on the patient’s preference, the perceived risk for early
When the duration of AF is longer than 48 hours or unclear, an recurrence of AF, and the duration of sinus rhythm between previous
alternative to 3 weeks of therapeutic anticoagulation before cardio- cardioversions. Treatment by cardioversion without daily antiarrhyth-
version is anticoagulation with heparin and transesophageal echo- mic drug therapy is acceptable if the episodes of AF are separated
cardiography to check for a left atrial thrombus. If no thrombi are by at least 6 months. Treatment with a rhythm-control drug is usually
seen, the patient can safely be cardioverted but still requires 4 weeks appropriate when AF recurs within a few months of cardioversion.
of therapeutic anticoagulation after cardioversion to prevent throm- The most realistic goal of antiarrhythmic drug therapy in patients
boembolism related to atrial stunning. The major clinical benefit of with persistent AF is to delay the onset of the next episode by at least
the transesophageal echocardiography–guided approach over the several months, not for several years. It is often appropriate to con-
conventional approach is that sinus rhythm is restored several weeks tinue therapy with a particular antiarrhythmic drug if recurrences of
sooner. When compared with the conventional approach, the trans- AF are limited to approximately one episode per year.
esophageal echocardiography–guided approach has not been found In patients with symptomatic paroxysmal AF, the aggressiveness
to reduce the risk for stroke or major bleeding or to affect the propor- with which a rhythm-control strategy is pursued should be dictated
tion of patients still in sinus rhythm at 8 weeks after cardioversion. by the frequency and severity of symptoms and how well antiarrhyth-
mic drug therapy is tolerated. Drug therapy is more likely to be judged
successful when patients are reminded that the goal of therapy is not
complete suppression of AF but a clinically meaningful reduction in
LONG-TERM MANAGEMENT the frequency, duration, and severity of episodes.
OF ATRIAL FIBRILLATION A pharmacologic rhythm-control strategy need not necessarily
consist of daily drug therapy. Episodic drug therapy (the “pill-in-the-
Pharmacologic Rate Control Versus pocket” approach) is useful for patients whose episodes of AF are
Rhythm Control relatively infrequent. Episodic drug therapy is a reasonable option for
Several randomized studies have compared a rate-control strategy patients who are clearly aware of the onset and termination of the
with a rhythm-control strategy in patients with AF. The largest study AF episodes and who have lone AF or only minimal structural heart
by far was the AFFIRM study, which consisted of 4060 patients with disease. A typical drug regimen consists of a class IC drug (flecainide
a mean age of 70 years who had AF for 6 hours to 6 months.35 At 5 or propafenone) plus a short-acting beta blocker (e.g., propranolol)
years of follow-up, the prevalence of sinus rhythm was 35% in the or calcium channel blocker (e.g., verapamil) for rate control. Many
rate-control arm and 63% in the rhythm-control arm. No significant patients with infrequent episodes prefer this approach because it
difference was noted between the two study arms in total mortality, eliminates the inconvenience, cost, and possible side effects of daily
stroke rate, or quality of life. The percentage of patients requiring prophylactic therapy. However, patients who are disabled by severe
hospitalization was significantly lower in the rate-control arm (73%) symptoms during AF may prefer daily prophylactic therapy even if
than in the rhythm-control arm (80%), and the incidence of adverse the episodes are infrequent.
drug effects such as torsades de pointes was also significantly lower Many patients with symptomatic AF also have asymptomatic epi-
in the rate-control arm (0.2% versus 0.8%). The authors of the AFFIRM sodes. Therefore, daily antithrombotic therapy to prevent thrombo-
study concluded that there is no survival advantage of a rhythm- embolic events is appropriate for all patients being treated for
control strategy over a rate-control strategy and that a rate-control recurrent AF, whether it is persistent or paroxysmal and whether a
strategy has advantages such as a lower probability of hospitalization rhythm-control or rate-control strategy is used. The choice of no
and adverse drug effects. therapy, an oral anticoagulant, aspirin, or the combination of aspirin
In a post hoc analysis of the AFFIRM study, the relationship between plus clopidogrel should be dictated by an analysis of risk factors and
sinus rhythm, treatment, and survival was determined by an drug tolerance.
on-treatment analysis instead of the intention-to-treat analysis used
in the original report.36 Sinus rhythm was found to be independently
associated with lower mortality (hazard ratio, 0.53), and antiarrhyth- Pharmacologic Rate Control
mic drug therapy was independently associated with increased An excessively rapid ventricular rate during AF often results in
mortality (hazard ratio, 1.49). Therefore, the potential benefit of main- uncomfortable symptoms and decreased effort tolerance and can
taining sinus rhythm with antiarrhythmic drugs was negated by the cause a tachycardia-induced cardiomyopathy if it is sustained for
adverse effects of the antiarrhythmic drug therapy. This suggested several weeks to months. Optimal heart rates during AF vary with age
that therapies that maintain sinus rhythm without major adverse and should be similar to the heart rates that a patient would have at
effects may have a beneficial effect on survival. a particular degree of exertion during sinus rhythm. Heart rate control
The results of the AFFIRM study should not be applied routinely to must be assessed both at rest and during exertion. At rest, the ideal
all patients with AF. The decision to pursue a rhythm-control strategy ventricular rate during AF is in the range of 60 to 75 beats/min. During
versus a rate-control strategy should be individualized, with several mild to moderate exertion (e.g., rapid walking), the target rate should
factors being taken into account, including the nature, frequency, and be 90 to 115 beats/min; and during strenuous exercise, the ideal rate
severity of symptoms; the length of time that AF has been present is in the range of 120 to 160 beats/min. Optimal assessment of the
continuously in patients with persistent AF; left atrial size; comorbid degree of heart rate control is provided by an ambulatory 24-hour
conditions; the response to previous cardioversions; age; the side Holter recording or an exercise test.
807
Oral agents available for long-term heart rate control in patients polymorphic ventricular tachycardia (torsades de pointes). Risk
with AF are digitalis, beta blockers, calcium channel antagonists, and factors for this type of proarrhythmia include female sex, left ventricu- 38
amiodarone. The first-line agents for rate control are beta blockers lar dysfunction, and hypokalemia. The risk for torsades de pointes

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


and the calcium channel antagonists verapamil and diltiazem. A appears to be much lower with dronedarone and amiodarone than
combination is often used to improve efficacy or to limit side effects with the other class III drugs. The ventricular proarrhythmia resulting
by allowing the use of smaller dosages of the individual drugs. In from class IC agents (flecainide and propafenone) is manifested as
patients with sinus node dysfunction and tachy-brady syndrome, use monomorphic ventricular tachycardia, sometimes associated with
of a beta blocker with intrinsic sympathomimetic activity (pindolol, widening of the QRS complex during sinus rhythm, but not with QT
acebutolol) may provide rate control without aggravating the sinus prolongation. Published studies indicate that the drugs most likely to
bradycardia. result in ventricular proarrhythmia are quinidine, flecainide, sotalol,
Digitalis may adequately control the rate at rest but often does not and dofetilide. In controlled studies, these agents increased the risk
provide adequate rate control during exertion. Its use is appropriate for ventricular tachycardia by a factor of 2 to 6.
in patients with systolic heart failure, in whom digitalis has been Adverse drug events resulting in discontinuation of drug therapy
shown to improve outcomes such as heart failure and all-cause hos- are fairly common with rhythm-control drugs. Withdrawal because
pitalization. In patients with the vagotonic form of paroxysmal AF, of adverse effects is most frequent with quinidine, disopyramide,
the vagotonic effect of digitalis may promote AF. Furthermore, in flecainide, sotalol, and amiodarone. A review of studies in which 32
patients without systolic heart failure, use of a digitalis glycoside may treatment arms received an antiarrhythmic drug for AF found that
have a deleterious effect on survival. In a large anticoagulation trial 10.4% of patients discontinued drug therapy because of an adverse
that compared warfarin with a direct thrombin inhibitor (SPORTIF drug event, most commonly gastrointestinal side effects and
III-IV), digitalis was found to be independently associated with a 53% neuropathy.39
higher risk for all-cause mortality.37 Although this was demonstrated The best options for drug therapy to suppress AF depend on the
by post hoc analysis and not by a randomized comparison of digitalis patient’s comorbid conditions. In patients with lone AF or minimal
versus placebo, the results are of enough concern to limit the use of heart disease (e.g., mild left ventricular hypertrophy), flecainide,
digitalis to patients with heart failure. propafenone, sotalol, and dronedarone are reasonable first-line
Amiodarone is much less frequently used for rate control than the drugs, and amiodarone and dofetilide can be considered if the first-
other negative dromotropic agents because of the risk for organ toxic- line agents are ineffective or not tolerated. In patients with substantial
ity associated with long-term therapy. Amiodarone may be an appro- left ventricular hypertrophy (left ventricular wall thickness >13 mm),
priate choice for rate control if the other agents are not tolerated or the hypertrophy heightens the risk for ventricular proarrhythmia, and
are ineffective. As an example, amiodarone would be an appropriate the safest choice for drug therapy is amiodarone. In patients with
choice for a patient with persistent AF, heart failure, and reactive coronary artery disease, several of the class I drugs have been found
airway disease who cannot tolerate either a calcium channel antago- to increase the risk for death, and the safest first-line options are
nist or a beta blocker and who has a rapid ventricular rate despite dofetilide, sotalol, and dronedarone, with amiodarone being reserved
treatment with digitalis. for use as a second-line agent. In patients with heart failure, several
Strict heart rate control can be difficult to achieve pharmacologi- antiarrhythmic drugs have been associated with increased mortality,
cally. The effects of a lenient rate-control strategy (resting rate <110 and the only two drugs known to have a neutral effect on survival
beats/min) and a strict rate-control strategy (resting heart rate <80 are amiodarone and dofetilide (see Chapter 35).
beats/min, rate during moderate exercise <110 beats/min) on out- At the time of its approval by the Food and Drug Administration,
comes were compared in randomized fashion in 614 patients with dronedarone was known to increase mortality in patients with class
persistent AF.38 The primary composite outcome was cardiovascular IV heart failure or those with a recent episode of decompensated
death, heart failure hospitalizations, stroke, embolism, major bleed- heart failure. After approval, the categories of patients in whom
ing episodes, and major arrhythmic events. The heart rate target was dronedarone is contraindicated expanded. A randomized study
achieved in 98% of patients in the lenient rate-control group versus (PALLAS) was terminated prematurely when it was found that drone-
67% in the strict rate-control group. The incidence of the primary darone increased the risk for heart failure, stroke, and cardiovascular
outcome at 3 years did not differ significantly between the lenient death in the following categories of patients: (1) 65 years or older
rate-control group (12.9%) and the strict rate-control group (14.9%). with permanent AF and either coronary artery disease, previous
The results suggest that strict rate control has no advantages over stroke, or symptomatic heart failure and (2) 75 years or older with
lenient rate control. However, the study did not present data on the hypertension and diabetes.40
severity of symptoms, exercise capacity, or left ventricular ejection
fraction, and follow-up was limited to 3 years. Strict rate control is still
often an appropriate goal for relief of symptoms, improvement in Rhythm Control with Agents Other Than
functional capacity, and avoidance of tachycardia-induced cardiomy- Antiarrhythmic Drugs
opathy during long-term follow-up. Experimental studies indicate that angiotensin-converting enzyme
(ACE) inhibitors and angiotensin receptor blockers (ARBs) have
favorable effects on electrical and structural remodeling (see
Pharmacologic Rhythm Control Chapter 33). This explains why ACE inhibitors and ARBs have been
The results of published studies on the efficacy of antiarrhythmic shown in some studies to prevent AF. However, other studies have
drugs for AF suggest that all the drugs available except amiodarone demonstrated that these agents do not prevent AF. For example, in a
have similar efficacy and are associated with a 50% to 60% reduction randomized clinical trial of the ARB valsartan versus placebo in 1442
in the odds of recurrent AF during 1 year of treatment. The one drug patients with structural heart disease and recurrent AF, the AF recur-
that stands out as having higher efficacy than the others is amioda- rence rate was approximately 50% in both study arms, and there was
rone. In studies that directly compared amiodarone with sotalol or no evidence that valsartan prevented AF.41 Therefore, at present the
class I drugs, amiodarone was 60% to 70% more effective in suppress- evidence is insufficient to support the use of ACE inhibitors and ARBs
ing AF. However, because of its risk for organ toxicity, amiodarone is for the sole purpose of preventing AF.
not appropriate first-line drug therapy for many patients with AF. Some evidence indicates that statins prevent AF, perhaps because
Because the efficacy of rhythm-control agents other than amioda- of their anti-inflammatory effects. A systematic review of 10 observa-
rone is in the same general range, selection of an antiarrhythmic drug tional studies demonstrated a 23% reduction in the relative risk for
to prevent AF is often dictated by the issues of safety and side effects. AF in patients treated with statins.42 However, a meta-analysis of six
Ventricular proarrhythmia from class IA agents (quinidine, pro- randomized clinical trials concluded that statins do not prevent AF,
cainamide, disopyramide) and class III agents (sotalol, dofetilide, except after open heart surgery.42 Therefore, the data available do not
dronedarone, amiodarone) is manifested as QT prolongation and support the use of statins for the prevention of AF.
808
Omega-3 polyunsaturated fatty acids (PUFAs) have an anti- catheter ablation is considered, particularly if the AF is persistent,
V inflammatory effect and can also have direct ion channel effects. because the efficacy of catheter ablation is lower for persistent AF
Several studies in which treatment with omega-3 PUFAs was initiated than for paroxysmal AF. The most appropriate candidates for catheter
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

at the time of cardioversion of AF demonstrated no prevention of ablation have symptomatic AF that is affecting their quality of life and
recurrent AF. However, omega-3 PUFAs did prevent recurrent AF after has not responded adequately to drug therapy. The ideal candidate
cardioversion of persistent AF in two prospective randomized studies has lone AF or only minimal structural heart disease. The recom-
in which patients were pretreated with 2 to 6 g/day of fish oil for 1 mendation for catheter ablation should be influenced by the esti-
month before cardioversion.43,44 Almost all patients in these studies mated probability of success, and the procedure is least likely to be
also received amiodarone or sotalol. The rationale for pretreatment successful if the left atrium is markedly dilated or if the AF has been
with the fish oil was to allow enough time for the omega-3 PUFAs persistent for more than 4 years.
to become incorporated into cell membranes and exert their ion Catheter ablation of AF is generally contraindicated in patients who
channel effects. These data suggest that fish oil can be helpful in have a left atrial thrombus or who cannot tolerate anticoagulation for
preventing recurrent AF when used in combination with an antiar- at least 6 to 8 weeks after ablation. Catheter ablation is also usually
rhythmic drug after cardioversion of persistent AF. inappropriate in asymptomatic individuals with a CHA 2DS2-VASc
score higher than 1 whose only motivation to undergo the procedure
is to eliminate the need for anticoagulation.
In a recent study, patients with symptomatic paroxysmal AF and
NONPHARMACOLOGIC MANAGEMENT no previous rhythm-control therapy were randomly assigned to
OF ATRIAL FIBRILLATION undergo catheter ablation or treatment with an antiarrhythmic drug.45
This study demonstrated that the cumulative AF burden at 2 years of
Pacing to Prevent Atrial Fibrillation follow-up did not differ significantly between the two groups. This
Randomized clinical trials comparing dual-chamber (DDD) pacing result validates the recommendation that catheter ablation be
with right ventricular pacing have concluded that atrial pacing pre- reserved for patients who have not responded adequately to treat-
vents AF. Studies suggest that the higher incidence of AF during ment with an antiarrhythmic drug. However, catheter ablation can
ventricular pacing than during DDD pacing may be at least partially be appropriate first-line therapy in some patients with AF: those
due to a proarrhythmic effect of ventricular pacing, not only to a younger than 35 years with symptomatic AF, those with sinus node
suppressive effect of atrial pacing. dysfunction in whom antiarrhythmic drug therapy is likely to create
Studies involving small numbers of patients have suggested that the need for a permanent pacemaker, and patients who express an
dual-site right atrial pacing or pacing of the interatrial septum near aversion to drug therapy.
the Bachmann bundle prevents AF. Although it is possible that these
atrial pacing techniques decrease the propensity for AF, the magni- Radiofrequency Catheter Ablation
tude of the effect appears to be minimal. The most commonly used energy to eliminate paroxysmal AF by
Some antibradycardia pacemakers are designed to prevent and catheter ablation is radiofrequency energy delivered through an
terminate AF. Pacing algorithms to prevent AF consist of atrial pacing irrigated-tip catheter. Radiofrequency energy is delivered point by
to prevent suppression of postextrasystolic pauses and acceleration point, typically in association with a three-dimensional electroana-
of the atrial pacing rate when repetitive premature atrial complexes tomic mapping system as a navigation guide and to create a visual
are sensed. When these pacing algorithms have been evaluated in record of the sites that have already been ablated. To improve ana-
rigorous fashion, they have been found to be ineffective or at best tomic accuracy, the electroanatomic map of the left atrium can be
minimally effective in reducing the AF burden. Antitachycardia merged with a computed tomography scan or magnetic resonance
pacing (ATP) to terminate AF consists of a burst of rapid atrial pacing image of the left atrium and pulmonary veins (Fig. 38-8) or with an
at the onset of AF. ATP may be useful for termination of atrial flutter ultrasound image generated by intracardiac echocardiography.
or atrial tachycardia, but it is rarely if ever effective for AF. Because of their important role in triggering and maintaining epi-
Because of insufficient evidence to support its use, atrial pacing is sodes of AF, almost all ablation strategies include electrical isolation
not indicated for prevention of AF in patients without bradycardia. In
patients with a bradycardia indication for a pacemaker and paroxys-
mal AF or recurrent episodes of persistent AF, the data available
clearly support the use of atrial-based pacing and programming to
minimize the amount of ventricular pacing.

Catheter Ablation of Atrial Fibrillation


Challenges of Ablating Atrial Fibrillation
Catheter ablation reliably and permanently eliminates several types of
arrhythmia, such as AV nodal reentrant tachycardia and accessory
pathway–mediated tachycardias36 (see Chapters 35 and 37). Success
rates greater than 95% are attainable when the arrhythmia substrate is
well defined, localized, and temporally stable. In contrast, the arrhyth-
mia substrate of AF is not well understood, is usually widespread, is
variable between patients, and may be progressive. Furthermore,
several factors that promote AF cannot be addressed simply by cath-
eter ablation, including comorbid conditions such as hypertension
and obstructive sleep apnea, structural remodeling of the atria, inflam-
matory factors, and genetic factors (see Chapter 8). Therefore, even
though late recurrences of AV nodal reentrant tachycardia or acces-
sory pathway conduction are very rare, AF can recur more than 2 or 3
years after an initially successful ablation procedure.
FIGURE 38-8 Electroanatomic map of the left atrium merged with a computed
tomography scan of the left atrium and pulmonary veins. The red and gray tags
Selection of Patients indicate the sites at which radiofrequency energy was delivered in the antrum of the
Given the limitations of catheter ablation of AF, it is usually appropri- pulmonary veins. LI = left inferior pulmonary vein; LS = left superior pulmonary vein;
ate to treat patients with at least one rhythm-control drug before RS = right superior pulmonary vein.
809

38
* *

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


I

V1

Abl
Ring catheter in LIPV

CS

250 msec

FIGURE 38-9 Tachycardia with a cycle length of 80 msec arising in a left inferior pulmonary vein. This tachycardia was responsible for generating AF. During radiofrequency
ablation in the antrum of this pulmonary vein, AF terminated (arrow) and converted to sinus rhythm when the pulmonary vein became electrically isolated. The sinus beats are
indicated with asterisks. The pulmonary vein tachycardia was still present inside the vein. Shown are leads I and V1, the electrograms recorded by the ablation catheter (Abl)
outside the left inferior pulmonary vein (LIPV) and by a ring catheter in the LIPV, and the coronary sinus electrograms (CS).

of the pulmonary veins (Fig. 38-9 and Video 38-1). Such isolation can of the localized sources in 86% of cases. At a median of 9 months of
be accomplished by either ostial ablation or wide-area ablation 1 to follow-up, 82% of patients were free of AF versus 45% of those in a
2 cm away from the ostia, in the antral regions of the pulmonary veins. control group who underwent conventional ablation. These early
Most of the data available indicate that wide-area ablation is more results suggest that focal impulse and rotor modulation can improve
effective than ostial ablation, probably because it also targets drivers outcomes of catheter ablation of AF.
that are in the antrum, outside the pulmonary vein itself.46 Triggers of Based on an extensive review of a large number of published
AF can also arise from other thoracic veins, such as the superior vena reports, the overall single-procedure success rate of radiofrequency
cava, coronary sinus, and the vein of Marshall. After the pulmonary catheter ablation of AF without antiarrhythmic drug therapy is 57%,
veins have been isolated, infusion of isoproterenol is helpful to deter- and the multiple-procedure success rate is 71%.39 Efficacy is strongly
mine whether any non–pulmonary vein triggers are present. influenced by the type of AF being ablated. For paroxysmal AF, a
Pulmonary vein isolation is often sufficient to eliminate paroxysmal single-procedure success rate of 60% to 75% is expected at experi-
AF but is usually insufficient for persistent AF. A variety of ablation enced centers, whereas for persistent AF, the single-procedure
strategies have been used for persistent AF after the pulmonary veins success rate is typically 50% or lower.
have been isolated: linear ablation across the left atrial roof, mitral The efficacy of radiofrequency catheter ablation of AF compares
isthmus, or cavotricuspid isthmus; ablation of CFAEs in the left atrium, favorably with that of antiarrhythmic drug therapy. In a meta-analysis
coronary sinus, or right atrium; various combinations of linear and of four prospective, randomized studies, radiofrequency catheter
CFAE ablation; and ablation of ganglionated plexuses.46 The endpoint ablation was found to result in AF-free survival significantly more
of catheter ablation of persistent AF is either completion of a prespeci- often than antiarrhythmic drug therapy was (76% versus 19%).47 In a
fied lesion set (in which case sinus rhythm is restored by cardiover- multicenter study of 112 patients with paroxysmal AF resistant to one
sion) or stepwise ablation until the AF converts to sinus rhythm. or more antiarrhythmic drugs, the patients were randomly assigned
A novel approach to ablation of AF is based on the hypothesis that to pulmonary vein isolation plus additional ablation at the operator’s
AF is sustained by localized sources, either rotors and/or focal discretion or antiarrhythmic drug therapy.48 The AF-free survival rate
impulses.5 Signal processing with proprietary software allowed iden- at 12 months was 89% in the ablation arm after a median of two
tification of focal impulses and rotors, which were then targeted for procedures per patient versus 23% in the drug arm.
radiofrequency ablation during ongoing episodes of AF. A mean of When the efficacy of catheter ablation of AF is evaluated, recur-
2.1 localized sources per patient were identified in 97% of 101 patients. rences of AF in the first 3 months after ablation are usually ignored.
Termination or slowing of AF was successfully achieved by ablation A 3-month blanking period excludes early recurrences that are
809.e1
VIDEO 38-1
Pulmonary vein (PV) isolation using image integration. A semi-trans- 38
lucent computed tomography “cast” of the left atrium appears in

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


orange, viewed first from an anterior perspective. As the movie runs,
black dots appear at sites sampled by a roving mapping catheter in
order to form a “shell” of the inner contours of the left atrium. The
image rotates to be viewed from the right side and then from the left.
Once this acquisition is completed, larger red circles appear at sites at
which radiofrequency energy is delivered to electrically isolate the PVs
by encircling their orifices with ablation lesions, starting with the left
PVs and ending with the right PVs. The image is rotated to allow
viewing of the process from different angles.
810
caused by a transient inflammatory response or incomplete lesion
V maturation.
Even in patients with symptomatic AF, postablation recurrences
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

can be asymptomatic. Therefore, accurate assessment of efficacy


requires monitoring for at least 7 days and preferably for 1 month with
a device capable of detecting asymptomatic episodes of AF. Ideally,
the monitoring should be performed at 6 and 12 months after the
procedure.
Most recurrences of AF after radiofrequency catheter ablation
occur within the first year of follow-up. However, recurrences con-
tinue to occur at a rate of approximately 5% to 6% per year for several
years after ablation.49 Ongoing comorbid conditions such as valvular
heart disease, obstructive sleep apnea, and hypertension can con-
tribute to late recurrences of AF. Another probable cause of late
recurrences of AF in patients who have undergone pulmonary vein
isolation is delayed recovery from radiofrequency-induced injury
leading to reconnection of the pulmonary veins.50
Atrial tachyarrhythmias that occur after catheter ablation of AF can
take the form of atrial tachycardia/flutter, which can be either focal
or reentrant. When the ablation strategy consists of only pulmonary
vein isolation, postablation focal atrial tachycardias are often attribut-
able to partial recovery of pulmonary vein conduction. The incidence
of reentrant atrial tachycardia/flutter after ablation is related to the
extent of ablation at atrial sites other than the pulmonary vein. When
extensive ablation is performed in the left atrium or in both atria in
an attempt to convert AF to sinus rhythm, atrial tachycardia/flutter
occurs during follow-up in more than 50% of patients. These arrhyth- FIGURE 38-10 Cryoballoon catheter positioned in the antrum of the right supe-
rior pulmonary vein. The balloon is inflated and there is no leakage of contrast mate-
mias do not respond well to antiarrhythmic drug therapy and fre- rial injected through the lumen of the cryoballoon catheter into the vein. A diagnostic
quently require another ablation procedure for elimination. ring catheter is positioned within the vein. Pulmonary vein isolation was achieved
The risk for a major complication after radiofrequency catheter with two 4-minute applications of cryoenergy through the balloon.
ablation of AF is reported to be 5% to 6%.39,51 In a large international
survey, the most common major complications were cardiac tampon-
ade (1.2%), pulmonary vein stenosis (1.3%), and cerebral thrombo-
embolism (0.94%).51 The risk for vascular injury is reported to be 1%
to 2%. The risk for an atrioesophageal fistula is probably less than who underwent endoscopy after cryoballoon ablation. Pulmonary
0.1%. Despite its rarity, this complication is of great concern because vein stenosis causing symptoms or requiring intervention is very infre-
it is often lethal. quent (0.2%), as are thromboembolic complications (0.6%). Cardiac
Large international surveys have reported the risk for a fatal com- tamponade has occurred in 0.6% of patients and vascular access
plication to be in the range of 0.05% to 0.1%.51,52 In a survey of 32,569 complications in approximately 2%.
patients who underwent catheter ablation of AF, the mortality rate
was 0.1%, and the most common causes of death were cardiac tam- Remote Magnetic Navigation
ponade (25% of deaths), stroke (16%), atrioesophageal fistula (16%), Two systems are currently available for remote navigation of a radio-
and pneumonia (16%).52 frequency ablation catheter. In one system, large magnets are posi-
A recently recognized complication of radiofrequency ablation tioned on each side of the patient, and small magnets embedded in
is silent cerebral ischemic lesions. Cerebral magnetic resonance the tip of the ablation catheter allow remote navigation by shifting
imaging demonstrated silent cerebral ischemic lesions in 14% of the magnetic field vectors. With the other system, an ablation cath-
patients in whom an irrigated-tip radiofrequency ablation catheter eter is navigated remotely by a robotic steerable sheath system. The
was used.53 The long-term clinical significance of these lesions is advantages of these systems are improved catheter stability, marked
unclear. reduction in exposure of the operator to radiation, and avoidance of
the technical challenges of manual catheter manipulation. The expe-
Cryoballoon Ablation rience to date with remote magnetic navigation indicates that the
In 2010, a cryoballoon catheter designed to isolate the pulmonary efficacy and safety of radiofrequency catheter ablation of AF
veins became widely available for use in the United States. In contrast are comparable to those of conventional radiofrequency catheter
to point-by-point radiofrequency ablation around pulmonary veins, ablation.55
the cryoballoon is designed to fit into the antrum of a pulmonary vein
and to create a circumferential ablative lesion via cryoenergy (Fig.
38-10). An advantage of cryoenergy over radiofrequency energy is Ablation of the Atrioventricular Node
that it is much less likely to cause pulmonary vein stenosis or esopha- Radiofrequency catheter ablation of the AV node results in complete
geal injury. Experienced operators can achieve complete pulmonary AV nodal blockade and substitutes a regular, paced rhythm for an
vein isolation acutely by using only the cryoballoon catheter in 98% irregular and rapid native rhythm. It is a useful strategy in patients
of patients.54 In patients with paroxysmal AF, 1-year freedom from AF who are symptomatic from AF because of a rapid ventricular rate that
is achieved in approximately 75% of patients.54 Randomized studies cannot be adequately controlled pharmacologically as a result of
have demonstrated no significant difference in freedom from AF at 1 either inefficacy of or intolerance to rate-control drugs and who either
year between cryoballoon ablation and radiofrequency ablation.54 are not good candidates for ablation of the AF or already have under-
The most frequent complication of cryoballoon ablation is phrenic gone an unsuccessful attempt at ablation of the AF.
nerve palsy, which has an incidence of approximately 7%.54 The In patients with AF and an uncontrolled ventricular rate, AV node
phrenic nerve palsy resolves within 1 year in almost all patients. In ablation improves the left ventricular ejection fraction in those with
one study, endoscopy showed esophageal ulcerations in 17% of 35 tachycardia-induced cardiomyopathy. AV node ablation has also
patients after cryoballoon ablation, but two other studies reported been shown to improve symptoms, quality of life, and functional
that there was no evidence of thermal esophageal injury in 81 patients capacity and to reduce the use of health care resources. There is no
811
evidence that AV node ablation is of benefit in patients whose ven-
tricular rate is well controlled by medications. SPECIFIC CLINICAL SYNDROMES 38
Disadvantages of AV node ablation are that it creates a life-long

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


need for ventricular pacing and does not restore AV synchrony. Postoperative Atrial Fibrillation
Although symptoms and functional capacity typically improve AF is common after open heart surgery and is reported to occur in
after AV node ablation in patients with AF and an uncontrolled ven- 25% to 40% of patients who undergo coronary artery bypass graft
tricular rate, some patients may not feel as well as during sinus surgery or valve replacement (see Chapter 80). It is associated with
rhythm. a twofold increase in the risk for postoperative stroke and is the most
AV node ablation is a technically simple procedure with an acute common reason for prolonged hospitalization. The risk for AF peaks
and long-term success rate of 98% or higher and a very low risk for on the second postoperative day. The pathogenesis of postoperative
complications. In patients with persistent AF, a ventricular pacemaker AF is multifactorial and probably involves adrenergic activation,
is implanted. A dual-chamber pacemaker is appropriate if the AF is inflammation, atrial ischemia, electrolyte disturbances, and genetic
paroxysmal. Most patients have a good clinical outcome with right factors. Several risk factors for AF after open heart surgery have been
ventricular pacing, but in those with borderline or depressed left identified, including age older than 70 years, history of previous AF,
ventricular function, biventricular pacing for cardiac resynchroniza- male sex, left ventricular dysfunction, left atrial enlargement, chronic
tion therapy is appropriate. In patients with ischemic or nonischemic lung disease, diabetes, and obesity.
cardiomyopathy and an ejection fraction from 30% or lower to 35%, The antiarrhythmic drugs that have been shown to decrease the
an ICD may be appropriate for primary prevention of sudden death. risk for postoperative AF are beta blockers, sotalol, and amiodarone.
However, a simple pacemaker without the ICD is often adequate for Amiodarone and sotalol decrease the risk for postoperative AF by
patients with a borderline ejection fraction (30% to 35%) and a rapid 50% to 65%, and beta blockers appear to be somewhat less effective,
ventricular rate because the ejection fraction is likely to improve to decreasing the risk by approximately 30%.
greater than 35% after the ventricular rate has been controlled by AV Hypomagnesemia is common after open heart surgery and can
node ablation. heighten the risk for AF. Magnesium administration immediately
preoperatively, perioperatively, or postoperatively is reported to
decreases the risk for postoperative AF by 20% to 40%.
Surgical Approaches to Atrial Fibrillation Right atrial or biatrial pacing using temporary electrodes attached
The most effective surgical procedure for AF is the “cut-and-sew” to the right and left atria is reported to reduce the risk for postopera-
maze procedure developed by Cox in 1987.56 This operation involves tive AF by 40%.
12 atrial incisions to isolate the pulmonary veins and create lines of The following agents have also been demonstrated in random-
block in the left atrium and right atrium. In addition, the left and right ized studies to reduce the risk for AF after open heart surgery: atorv-
atrial appendages are excised. Rates of long-term freedom from AF astatin, which decreases the risk by approximately 60%;
after the Cox maze procedure are reported to range from 70% to 95%, hydrocortisone, which decreases the risk by approximately 35%60;
but 10% to 35% of patients still require antiarrhythmic drug therapy. and colchicine, which decreases the risk by 45%.61 The main mecha-
The efficacy of the Cox maze procedure is lower in patients with a nism by which these agents prevent AF is probably an anti-
very large left atrium or with persistent AF for many years. The Cox inflammatory effect. Omega-3 PUFAs, which also have an
maze procedure has not been widely performed because it requires anti-inflammatory effect, reduced the risk for postoperative AF by
cardiopulmonary bypass, is technically difficult, and is associated 43% in one randomized study62 but did not reduce the risk in another
with a mortality rate of approximately 1% to 2%. randomized study.63
A large variety of surgical ablation tools have been developed to
simplify the classic Cox maze procedure. These tools allow the
surgeon to substitute an ablation line for a surgical incision. Several Wolff-Parkinson-White Syndrome
different types of energy have been used for surgical ablation: radio- Patients with Wolff-Parkinson-White syndrome and an accessory
frequency energy, cryoenergy, microwave, laser, and high-intensity pathway with a short refractory period can experience a very rapid
focused ultrasound. The tool that most consistently produces trans- ventricular rate during AF (see Chapters 35 and 37). Ventricular
mural ablation lines is a clamp device intended to isolate the pulmo- rates higher than 250 to 300 beats/min can result in loss of conscious-
nary veins with bipolar radiofrequency energy. ness or precipitate ventricular fibrillation and cardiac arrest. Patients
Various surgical ablation strategies have been used, including pul- with Wolff-Parkinson-White syndrome who have AF with a rapid ven-
monary vein isolation, left atrial ablation, and the Cox maze lesion tricular rate should undergo transthoracic cardioversion if they are
set. The left atrial appendage is usually excluded and ganglionic hemodynamically unstable. If the patient is hemodynamically stable,
plexuses are often also ablated. In patients who do not require con- intravenous procainamide or ibutilide can be used for pharmaco-
comitant coronary artery bypass grafting or valve repair or replace- logic cardioversion. Procainamide may be preferable to ibutilide
ment, surgical ablation is typically performed via a thorascopic, because it blocks accessory pathway conduction and slows the ven-
minimally invasive epicardial approach. Single-procedure success tricular rate before AF has converted to sinus rhythm. Digitalis and
rates at 1 year of follow-up have ranged from 65% to 86%.57,58 calcium channel antagonists are contraindicated in patients with
Radiofrequency catheter ablation of AF was compared with surgi- Wolff-Parkinson-White syndrome and AF. These agents selectively
cal ablation in a randomized study that selected patients with drug- block conduction in the AV node and can result in acceleration of
refractory AF who had hypertension and left atrial dilation or who conduction through the accessory pathway.
had already undergone unsuccessful catheter ablation.59 Freedom The preferred therapy for patients with Wolff-Parkinson-White syn-
from AF at 1 year was significantly higher in the surgical group drome and AF with a rapid ventricular rate is catheter ablation of the
(65.6%) than in the catheter ablation group (36.5%). However, sub- accessory pathway. The efficacy of catheter ablation is 95% for most
stantial procedural adverse events also occurred significantly more types of accessory pathways, and the risk for a major complication
often with surgical ablation (23%) than with catheter ablation (3.2%). is very low. AF typically no longer recurs after successful accessory
The most common complications of surgical ablation were hemotho- pathway ablation, probably because AF in Wolff-Parkinson-White
rax and bradycardia requiring pacemaker implantation. syndrome is often induced by tachycardia and is a result of AV recip-
Surgical therapy for AF is appropriate as a concomitant procedure rocating tachycardia.
in patients undergoing open heart surgery for coronary artery disease
or valvular disease. A stand-alone surgical procedure for AF is an
option for patients who have not had a successful outcome from Heart Failure
catheter ablation, who are not good candidates for catheter ablation, AF is a common arrhythmia in patients with heart failure, with a
or who prefer a surgical procedure over catheter ablation. prevalence reported to range from 10% in patients with New York
812
Heart Association functional class I up to 50% in patients with class FUTURE PERSPECTIVES
V IV (see Chapter 25). AF may be the cause of heart failure in patients
with nonischemic cardiomyopathy and AF with a rapid ventricular The ideal antiarrhythmic drug to prevent AF would affect the atrium
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

rate. In patients with structural heart disease and preexisting left only, thereby eliminating the potential for ventricular proarrhythmia.
ventricular dysfunction, AF can worsen the heart failure. The deleteri- Such drugs are under development and may improve the safety and
ous hemodynamic effects of AF are mediated by a rapid rate or efficacy of pharmacologic therapy for AF. It is likely that drugs that
irregular ventricular rate and loss of AV synchrony. modify a single channel will not be as effective as those with multiple
The most appropriate rate-control drugs in patients with heart actions, and it is possible that targeting of non–channel-related func-
failure are digitalis and beta blockers. If necessary, amiodarone can tions such as the development of fibrosis will prove useful.
also be used for rate control. The only rhythm-control drugs safe to In the past few years, significant progress has been made in the
use in patients with heart failure are amiodarone and dofetilide. field of catheter ablation of AF, but there is still much room for
These are the only two rhythm-control drugs that have been demon- improvement in efficacy and procedure duration. The failure to
strated to not increase the risk for death in patients with heart failure. create enduring pulmonary vein isolation often accounts for recur-
As in other patients with AF, the decision to pursue a rate-control rences of AF in patients with paroxysmal AF. The development of new
or rhythm-control strategy in patients with heart failure should be tools for catheter ablation, such as radiofrequency ablation catheters
individualized. In a multicenter study, patients with AF, heart failure, that sense tissue contact force, might improve the ability to safely
and a mean left ventricular ejection fraction of 27% were randomly create transmural lesions, thereby limiting the need for repeated
assigned to a rate-control strategy (most commonly digitalis and beta ablation procedures. In patients with persistent AF, a better under-
blockers) or a rhythm-control strategy (most often amiodarone).64 At standing of AF mechanisms could result in more efficient and suc-
3 years of follow-up, no significant differences were noted in all-cause cessful ablation strategies. The recent demonstration of localized
mortality, cardiovascular mortality, or worsening heart failure, but the sources of AF (focal impulses and/or atrial rotor) by computed signal
hospitalization rate was higher in the rhythm-control group. This analysis in humans represents an important step in this direction.5
study demonstrated no beneficial effect of a rhythm-control strategy Several studies have shown that a rhythm-control strategy in
on outcomes in patients with heart failure. However, endpoints such patients with AF provides no advantages in outcomes over a rate-
as ejection fraction and functional capacity were not examined in control strategy. The results of these studies were most likely influ-
the study, and many patients in the rhythm-control arm continued to enced by the suboptimal safety and efficacy of the drugs used for
have AF. It should be noted that the study compared two treatment rhythm control.
strategies, not sinus rhythm versus AF with a controlled ventricular To date, no randomized trials have demonstrated that catheter
rate. The study did not rule out the possibility that sinus rhythm has ablation of AF improves outcomes such as stroke or survival. The
advantages over AF with a controlled ventricular rate in patients with ongoing trial CABANA (Catheter Ablation versus Antiarrhythmic
heart failure. Drug Therapy for Atrial Fibrillation) has a primary endpoint of mortal-
In another randomized study, patients with AF, heart failure, and ity and secondary endpoints of cardiovascular mortality and stroke.
an ejection fraction lower than 40% were randomly assigned to If this study shows improved outcomes with AF ablation, it will
rhythm control by catheter ablation or rate control by AV node strengthen the case for rhythm control by ablation.
ablation plus a biventricular pacemaker.65 The rate of freedom
from AF at 6 months in the AF ablation arm was 71% in the absence References
of antiarrhythmic drug therapy. The mean ejection fraction improved Epidemiology of Atrial Fibrillation
1. Roger VL, Go AS, Lloyd-Jones DM, et al: Heart disease and stroke statistics—2012 update: A
from approximately 27% to 35% in the AF ablation group and remained report from the American Heart Association. Circulation 125:e2, 2012.
unchanged in the AV node ablation group. Functional capacity 2. Gami AS, Hodge DO, Herges RM, et al: Obstructive sleep apnea, obesity, and the risk of inci-
dent atrial fibrillation. J Am Coll Cardiol 49:565, 2007.
and quality of life also improved significantly in the AF ablation 3. Ahlehoff O, Gislason GH, Jorgensen CH, et al: Psoriasis and risk of atrial fibrillation and
group, but not in the AV node ablation group. The failure of AV node ischaemic stroke: A Danish nationwide cohort study. Eur Heart J 33:2054, 2012.
ablation to improve these parameters is attributable to the fact 4. Miyasaka Y, Barnes ME, Gersh BJ, et al: Secular trends in incidence of atrial fibrillation in
Olmsted County, Minnesota, 1980 to 2000, and implications on the projections for future
that the patients had already achieved adequate rate control with prevalence. Circulation 114:119, 2006.
drug therapy.
These results suggest that attempts at restoration of sinus rhythm Mechanisms of Atrial Fibrillation
5. Narayan SM, Krummen DE, Shivkumar K, et al: Treatment of atrial fibrillation by the ablation
are worthwhile in patients with heart failure and that catheter abla- of localized sources: CONFIRM (Conventional Ablation For Atrial Fibrillation with or without
tion of AF should be considered if sinus rhythm is not maintained by Focal Impulse and Rotor Modulation) trial. J Am Coll Cardiol 60:628, 2012.
amiodarone or dofetilide. A rate-control strategy is appropriate for
patients who do not respond adequately to amiodarone or dofetilide Genetic Factors
6. Tsai CT, Lai LP, Hwang JJ, Lin JL, Chiang FT: Molecular genetics of atrial fibrillation. J Am Coll
and either are not suitable candidates for catheter ablation of AF or Cardiol 52:241, 2008.
have had an unsuccessful outcome from ablation. AV node ablation
should be reserved for patients whose ventricular rate during AF is Prevention of Thromboembolic Complications
7. Lip GY, Edwards SJ: Stroke prevention with aspirin, warfarin and ximelagatran in patients
not adequately controlled by drug therapy. Because left ventricular with non-valvular atrial fibrillation: A systematic review and meta-analysis. Thromb Res
dysfunction and heart failure can be aggravated by right ventricular 118:321, 2006.
8. Fuster V, Ryden LE, Cannom DS, et al: 2011 ACCF/AHA/HRS focused updates incorporated
pacing, biventricular pacing should be performed after AV node abla- into the ACC/AHA/ESC 2006 guidelines for the management of patients with atrial fibrilla-
tion. The decision to implant a biventricular pacemaker versus a tion: A report of the American College of Cardiology Foundation/American Heart Associa-
tion Task Force on Practice Guidelines. Circulation 123:e269, 2011.
biventricular ICD is based on clinical judgment. If it seems likely that 9. Friberg L, Rosenqvist M, Lip GY: Evaluation of risk stratification schemes for ischaemic stroke
the ejection fraction will remain less than 30% to 35% after optimal and bleeding in 182 678 patients with atrial fibrillation: The Swedish Atrial Fibrillation Cohort
heart rate control, a biventricular ICD is appropriate for primary pre- Study. Eur Heart J 33:1500, 2012.
10. Lip GY: Implications of the CHA(2)DS(2)-VASc and HAS-BLED scores for thromboprophy-
vention of sudden cardiac death. laxis in atrial fibrillation. Am J Med 124:111, 2011.
11. Go AS, Fang MC, Udaltsova N, et al: Impact of proteinuria and glomerular filtration rate on
risk of thromboembolism in atrial fibrillation: The Anticoagulation and Risk Factors in Atrial
Fibrillation (ATRIA) study. Circulation 119:1363, 2009.
Pregnancy 12. Akar JG, Jeske W, Wilber DJ: Acute onset human atrial fibrillation is associated with local
cardiac platelet activation and endothelial dysfunction. J Am Coll Cardiol 51:1790, 2008.
New-onset AF is rare during pregnancy (see Chapter 78), and when 13. Healey JS, Connolly SJ, Gold MR, et al: Subclinical atrial fibrillation and the risk of stroke.
it does occur, it is usually in the setting of underlying congenital or N Engl J Med 366:120, 2012.
valvular heart disease, thyrotoxicosis, or electrolyte abnormalities. In 14. Lip GY, Frison L, Halperin JL, Lane DA: Comparative validation of a novel risk score for
predicting bleeding risk in anticoagulated patients with atrial fibrillation: The HAS-BLED
women with paroxysmal AF before pregnancy, the frequency of epi- (hypertension, abnormal renal/liver function, stroke, bleeding history or predisposition,
sodes may or may not increase during pregnancy. Specific recom- labile INR, elderly, drugs/alcohol concomitantly) score. J Am Coll Cardiol 57:173, 2011.
15. Pisters R, Lane DA, Nieuwlaat R, et al: A novel user-friendly score (HAS-BLED) to assess
mendations for the management of AF during pregnancy are provided 1-year risk of major bleeding in patients with atrial fibrillation: The Euro Heart Survey. Chest
in the Guidelines for Atrial Fibrillation. 138:1093, 2010.
813
16. Olesen JB, Lip GY, Hansen ML, et al: Validation of risk stratification schemes for predicting 42. Liu T, Li L, Korantzopoulos P, et al: Statin use and development of atrial fibrillation: A system-
stroke and thromboembolism in patients with atrial fibrillation: Nationwide cohort study. atic review and meta-analysis of randomized clinical trials and observational studies. Int J
BMJ 342:d124, 2011. Cardiol 126:160, 2008. 38
17. Friberg L, Rosenqvist M, Lip GY: Net clinical benefit of warfarin in patients with atrial fibrilla- 43. Nodari S, Triggiani M, Campia U, et al: N-3 polyunsaturated fatty acids in the prevention

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


tion: A report from the Swedish Atrial Fibrillation Cohort Study. Circulation 125:2298, 2012. of atrial fibrillation recurrences after electrical cardioversion: A prospective, randomized
18. Wann LS, Curtis AB, January CT, et al: 2011 ACCF/AHA/HRS focused update on the manage- study. Circulation 124:1100, 2011.
ment of patients with atrial fibrillation (updating the 2006 guideline): A report of the Ameri- 44. Kumar S, Sutherland F, Morton JB, et al: Long-term omega-3 polyunsaturated fatty acid sup-
can College of Cardiology Foundation/American Heart Association Task Force on Practice plementation reduces the recurrence of persistent atrial fibrillation after electrical cardiover-
Guidelines. J Am Coll Cardiol 57:223, 2011. sion. Heart Rhythm 9:483, 2012.
19. Camm AJ, Lip GY, De Caterina R, et al: 2012 focused update of the ESC guidelines for the
management of atrial fibrillation: An update of the 2010 ESC guidelines for the management
of atrial fibrillation—Developed with the special contribution of the European Heart Rhythm Nonpharmacologic Management of Atrial Fibrillation
Association. Eur Heart J 14:1385, 2012. 45. Nielsen JC, Johannsessen A, Raatikainen P, et al: Radiofrequency ablation as initial therapy
20. Connolly SJ, Pogue J, Hart RG, et al: Effect of clopidogrel added to aspirin in patients with in paroxysmal atrial fibrillation. N Engl J Med 367:1587, 2012.
atrial fibrillation. N Engl J Med 360:2066, 2009. 46. Calkins H, Kuck KH, Cappato R, et al: 2012 HRS/EHRA/ECAS expert consensus statement
21. Rose AJ, Ozonoff A, Henault LE, Hylek EM: Warfarin for atrial fibrillation in community-based on catheter and surgical ablation of atrial fibrillation: Recommendations for patient selec-
practise. J Thromb Haemost 6:1674, 2008. tion, procedural techniques, patient management and follow-up, definitions, endpoints, and
22. Mant J, Hobbs FD, Fletcher K, et al: Warfarin versus aspirin for stroke prevention in an elderly research trial design. J Interv Card Electrophysiol 33:171, 2012.
community population with atrial fibrillation (the Birmingham Atrial Fibrillation Treatment 47. Noheria A, Kumar A, Wylie JV Jr, Josephson ME: Catheter ablation vs antiarrhythmic drug
of the Aged study, BAFTA): A randomised controlled trial. Lancet 370:493, 2007. therapy for atrial fibrillation: A systematic review. Arch Intern Med 168:581, 2008.
23. Klein TE, Altman RB, Eriksson N, et al: Estimation of the warfarin dose with clinical and 48. Jais P, Cauchemez B, Macle L, et al: Catheter ablation versus antiarrhythmic drugs for atrial
pharmacogenetic data. N Engl J Med 360:753, 2009. fibrillation: The A4 study. Circulation 118:2498, 2008.
24. Anderson JL, Horne BD, Stevens SM, et al: Randomized trial of genotype-guided versus stand- 49. Weerasooriya R, Khairy P, Litalien J, et al: Catheter ablation for atrial fibrillation: Are results
ard warfarin dosing in patients initiating oral anticoagulation. Circulation 116:2563, 2007. maintained at 5 years of follow-up? J Am Coll Cardiol 57:160, 2011.
25. Connolly SJ, Ezekowitz MD, Yusuf S, et al: Dabigatran versus warfarin in patients with atrial 50. Kowalski M, Grimes MM, Perez FJ, et al: Histopathologic characterization of chronic radiofre-
fibrillation. N Engl J Med 361:1139, 2009. quency ablation lesions for pulmonary vein isolation. J Am Coll Cardiol 59:930, 2012.
26. Patel MR, Mahaffey KW, Garg J, et al: Rivaroxaban versus warfarin in nonvalvular atrial fibril- 51. Cappato R, Calkins H, Chen SA, et al: Updated worldwide survey on the methods, efficacy,
lation. N Engl J Med 365:883, 2011. and safety of catheter ablation for human atrial fibrillation. Circ Arrhythm Electrophysiol
27. Granger CB, Alexander JH, McMurray JJ, et al: Apixaban versus warfarin in patients with 3:32, 2010.
atrial fibrillation. N Engl J Med 365:981, 2011. 52. Cappato R, Calkins H, Chen SA, et al: Prevalence and causes of fatal outcome in catheter
28. Eerenberg ES, Kamphuisen PW, Sijpkens MK, et al: Reversal of rivaroxaban and dabigatran ablation of atrial fibrillation. J Am Coll Cardiol 53:1798, 2009.
by prothrombin complex concentrate: A randomized, placebo-controlled, crossover study in 53. Gaita F, Caponi D, Pianelli M, et al: Radiofrequency catheter ablation of atrial fibrillation:
healthy subjects. Circulation 124:1573, 2011. A cause of silent thromboembolism? Magnetic resonance imaging assessment of cerebral
29. Wann LS, Curtis AB, Ellenbogen KA, et al: 2011 ACCF/AHA/HRS focused update on the man- thromboembolism in patients undergoing ablation of atrial fibrillation. Circulation 122:1667,
agement of patients with atrial fibrillation (update on dabigatran): A report of the Ameri- 2010.
can College Of Cardiology Foundation/American Heart Association Task Force on Practice 54. Andrade JG, Khairy P, Guerra PG, et al: Efficacy and safety of cryoballoon ablation for atrial
Guidelines. Circulation 123:1144, 2011. fibrillation: A systematic review of published studies. Heart Rhythm 8:1444, 2011.
30. Nagarakanti R, Ezekowitz MD, Oldgren J, et al: Dabigatran versus warfarin in patients with 55. Di Biase L, Wang Y, Horton R, et al: Ablation of atrial fibrillation utilizing robotic catheter
atrial fibrillation: An analysis of patients undergoing cardioversion. Circulation 123:131, 2011. navigation in comparison to manual navigation and ablation: Single-center experience.
31. Lakkireddy D, Reddy YM, Di Biase L, et al: Feasibility and safety of dabigatran versus war- J Cardiovasc Electrophysiol 20:1328, 2009.
farin for periprocedural anticoagulation in patients undergoing radiofrequency ablation for 56. Cox JL, Boineau JP, Schuessler RB, et al: Electrophysiologic basis, surgical development, and
atrial fibrillation: Results from a multicenter prospective registry. J Am Coll Cardiol 59:1168, clinical results of the maze procedure for atrial flutter and atrial fibrillation. Adv Card Surg
2012. 6:1, 1995.
32. Kanderian AS, Gillinov AM, Pettersson GB, et al: Success of surgical left atrial appendage 57. Han FT, Kasirajan V, Kowalski M, et al: Results of a minimally invasive surgical pulmonary
closure: Assessment by transesophageal echocardiography. J Am Coll Cardiol 52:924, 2008. vein isolation and ganglionic plexi ablation for atrial fibrillation: Single-center experience
33. Holmes DR, Reddy VY, Turi ZG, et al: Percutaneous closure of the left atrial appendage versus with 12-month follow-up. Circ Arrhythm Electrophysiol 2:370, 2009.
warfarin therapy for prevention of stroke in patients with atrial fibrillation: A randomised 58. Krul SP, Driessen AH, van Boven WJ, et al: Thoracoscopic video-assisted pulmonary vein
non-inferiority trial. Lancet 374:534, 2009. antrum isolation, ganglionated plexus ablation, and periprocedural confirmation of ablation
34. Reddy VY, Holmes D, Doshi SK, et al: Safety of percutaneous left atrial appendage closure: lesions: First results of a hybrid surgical-electrophysiological approach for atrial fibrillation.
Results from the Watchman Left Atrial Appendage System For Embolic Protection In Patients Circ Arrhythm Electrophysiol 4:262, 2011.
With AF (PROTECT AF) clinical trial and the continued access registry. Circulation 123:417, 2011. 59. Boersma LV, Castella M, van Boven W, et al: Atrial fibrillation catheter ablation versus surgi-
cal ablation treatment (FAST): A 2-center randomized clinical trial. Circulation 125:23, 2012.

Long-Term Management of Atrial Fibrillation


35. Wyse DG, Waldo AL, DiMarco JP, et al: A comparison of rate control and rhythm control in Specific Clinical Syndromes
patients with atrial fibrillation. N Engl J Med 347:1825, 2002. 60. Halonen J, Halonen P, Jarvinen O, et al: Corticosteroids for the prevention of atrial fibrillation
36. Corley SD, Epstein AE, DiMarco JP, et al: Relationships between sinus rhythm, treatment, and after cardiac surgery: A randomized controlled trial. JAMA 297:1562, 2007.
survival in the Atrial Fibrillation Follow-Up Investigation of Rhythm Management (AFFIRM) 61. Imazio M, Brucato A, Ferrazzi P, et al: Colchicine reduces postoperative atrial fibrillation:
study. Circulation 109:1509, 2004. Results of the Colchicine for the Prevention of the Postpericardiotomy Syndrome (COPPS)
37. Gjesdal K, Feyzi J, Olsson SB: Digitalis: A dangerous drug in atrial fibrillation? An analysis of atrial fibrillation substudy. Circulation 124:2290, 2011.
the SPORTIF III and V data. Heart 94:191, 2008. 62. Heidt MC, Vician M, Stracke SK, et al: Beneficial effects of intravenously administered n-3
38. Van Gelder IC, Groenveld HF, Crijns HJ, et al: Lenient versus strict rate control in patients with fatty acids for the prevention of atrial fibrillation after coronary artery bypass surgery: A
atrial fibrillation. N Engl J Med 362:1363, 2010. prospective randomized study. Thorac Cardiovasc Surg 57:276, 2009.
39. Calkins H, Reynolds MR, Spector P, et al: Treatment of atrial fibrillation with antiarrhythmic 63. Saravanan P, Bridgewater B, West AL, et al: Omega-3 fatty acid supplementation does not
drugs or radiofrequency ablation: Two systematic literature reviews and meta-analyses. Circ reduce risk of atrial fibrillation after coronary artery bypass surgery: A randomized, double-
Arrhythm Electrophysiol 2:349, 2009. blind, placebo-controlled clinical trial. Circ Arrhythm Electrophysiol 3:46, 2010.
40. Connolly SJ, Camm AJ, Halperin JL, et al: Dronedarone in high-risk permanent atrial fibrilla- 64. Roy D, Talajic M, Nattel S, et al: Rhythm control versus rate control for atrial fibrillation and
tion. N Engl J Med 365:2268, 2011. heart failure. N Engl J Med 358:2667, 2008.
41. Disertori M, Latini R, Barlera S, et al: Valsartan for prevention of recurrent atrial fibrillation. N 65. Khan MN, Jais P, Cummings J, et al: Pulmonary-vein isolation for atrial fibrillation in patients
Engl J Med 360:1606, 2009. with heart failure. N Engl J Med 359:1778, 2008.

GUIDELINES Class IIa: weight of evidence or opinion in favor of usefulness/efficacy


Class IIb: usefulness or efficacy less well established by evidence or
Atrial Fibrillation opinion
Class III: conditions for which there is evidence and/or general agree-
Fred Morady and Douglas P. Zipes ment that the test is not useful or effective and in some cases may
be harmful
Recent updates have been incorporated into the comprehensive Level A: recommendations derived from data from multiple random-
guidelines for the management of atrial fibrillation (AF) published ized clinical trials
in 2006 by the American College of Cardiology/American Heart Level B: recommendations derived from a single randomized trial or
Association (ACC/AHA) Task Force on Practice Guidelines and the nonrandomized studies
European Society of Cardiology Committee for Practice Guidelines.1 Level C: recommendations based on the consensus opinion of
The following classification system was used for recommendations experts
and for the level of evidence on which the recommendations are The guidelines do not necessarily define the standard of care.
based: Management decisions must be individualized on the basis of the
Class I: conditions for which there is evidence and/or general agree- particular circumstances of a patient, and in some situations, devia-
ment that the test is useful and effective tion from the guidelines may be appropriate.
814
CLASSIFICATION OF ATRIAL FIBRILLATION Wolff-Parkinson-White syndrome, hyperthyroidism, pregnancy, hyper-
V trophic cardiomyopathy, and pulmonary disease.
The terminology used to classify AF in the guidelines is as follows: An important aspect in the management of patients with AF that
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

paroxysmal AF is defined as episodes of AF that last less than 7 days, is not specifically addressed in the guidelines is how to decide on a
persistent AF is defined as AF that lasts more than 7 days, and long- rate-control strategy versus a rhythm-control strategy. Several clinical
standing AF refers to AF that has been persistent for more than 1 year. trials have demonstrated that pharmacologic rhythm-control and
These designations are not altered by termination of AF via drug rate-control strategies result in similar outcomes, even in patients
therapy or electrical cardioversion. AF that is resistant to electrical with AF who have left ventricular dysfunction and heart failure. On
cardioversion is referred to as permanent AF. AF is considered recur- the other hand, a randomized trial of left atrial radiofrequency cath-
rent after two or more episodes have occurred. eter ablation versus atrioventricular (AV) node ablation and biven-
Some patients with paroxysmal AF occasionally have episodes that tricular pacing demonstrated significantly greater improvement in left
are persistent, and vice versa. In this event the AF should be catego- ventricular function, exercise capacity, and quality of life in patients
rized on the basis of the predominant form. Lone AF refers to AF in with heart failure who were treated by ablation. It is possible that the
patients younger than 60 years who do not have structural heart side effects from the drugs used for rhythm control offset the benefits
disease or hypertension. AF that is secondary to acute myocardial of sinus rhythm. Specific recommendations regarding rhythm-control
infarction, cardiac surgery, pericarditis, myocarditis, hyperthyroid- versus rate-control strategies are difficult to provide because the deci-
ism, or an acute pulmonary process is considered separately because sion must be individualized on the basis of several factors, including
the AF often resolves after treatment of the underlying disorder. age, symptom severity, functional limitations, patient preference,
comorbid conditions, sinus node function, and response to drug
therapy.
MANAGEMENT OF ATRIAL FIBRILLATION
The guidelines address five aspects of the management of AF: phar- Pharmacologic Rate Control During Atrial
macologic rate control; prevention of thromboembolic complica- Fibrillation (Table 38G-1)
tions; cardioversion; maintenance of sinus rhythm; and special In addition to specific recommendations on the use of particular
considerations, including postoperative AF, myocardial infarction, drugs for control of the ventricular rate, the guidelines recommend

TABLE 38G-1 ACC/AHA Recommendations for Pharmacologic Rate Control of Atrial Fibrillation
LEVEL OF
CLASS INDICATION EVIDENCE
Class I Measurement of the heart rate at rest and control of the rate with pharmacologic agents (in most cases either a B
(indicated) beta blocker or nondihydropyridine calcium channel antagonist) are recommended for patients with persistent or
permanent AF
In the absence of preexcitation, intravenous administration of beta blockers (esmolol, metoprolol, or propranolol) B
or nondihydropyridine calcium channel antagonists (verapamil, diltiazem) is recommended to slow the ventricular
response to AF in the acute setting, with caution being exercised in patients with hypotension or heart failure
Intravenous administration of digoxin or amiodarone is recommended to control the heart rate in patients with AF and B
heart failure who do not have an accessory pathway
In patients who experience symptoms related to AF during activity, the adequacy of heart rate control should be C
assessed during exercise, with pharmacologic treatment being adjusted as necessary to keep the rate in the
physiologic range
Digoxin is effective after oral administration to control the heart rate at rest in patients with AF and is indicated for C
patients with heart failure or left ventricular dysfunction and for sedentary individuals
Class IIa A combination of digoxin and either a beta blocker or nondihydropyridine calcium channel antagonist is reasonable B
(reasonable) to control the heart rate both at rest and during exercise in patients with AF. The choice of medication should be
individualized and the dose modulated to avoid bradycardia
It is reasonable to use ablation of the AV node or accessory pathway to control the heart rate when pharmacologic B
therapy is insufficient or associated with side effects
Intravenous amiodarone can be useful to control heart rate in patients with AF when other measures are unsuccessful C
or contraindicated
When electrical cardioversion is not necessary in patients with AF and an accessory pathway, intravenous procainamide C
or ibutilide is a reasonable alternative
Class IIb When the ventricular rate cannot be adequately controlled both at rest and during exercise in patients with AF by a C
(may be beta blocker, nondihydropyridine calcium channel antagonist, or digoxin, alone or in combination, oral amiodarone
considered) may be administered to control the heart rate
Intravenous procainamide, disopyramide, ibutilide, or amiodarone may be considered for hemodynamically stable B
patients with AF involving conduction over an accessory pathway
When the rate cannot be controlled with pharmacologic agents or tachycardia-mediated cardiomyopathy is suspected, C
catheter-directed ablation of the AV node may be considered in patients with AF to control the heart rate
Class III (not Strict rate control (<80 beats/min at rest or <110 beats/min during a 6-minute walk) is not more beneficial than a B
indicated ) resting rate of <110 beats/min in asymptomatic patients with persistent AF and an ejection fraction >40%, although
uncontrolled tachycardia can lead to reversible left ventricular dysfunction over time
Digitalis should not be used as the sole agent to control the rate of ventricular response in patients with paroxysmal AF B
Catheter ablation of the AV node should not be attempted without a prior trial of medication to control the C
ventricular rate in patients with AF
In patients with decompensated heart failure and AF, intravenous administration of a nondihydropyridine calcium C
channel antagonist may exacerbate the hemodynamic compromise and is not recommended
Intravenous administration of digitalis glycosides or nondihydropyridine calcium channel antagonists to patients with C
AF and a preexcitation syndrome may paradoxically accelerate the ventricular response and is not recommended
815
that the effects of drug therapy on ventricular rate be measured at AF except those who have lone AF or contraindications. An updated
rest and during exercise to ensure adequate heart rate control. The recommendation as of 2011 is that the direct thrombin inhibitor dabi- 38
criteria used for rate control are rates of 60 to 80 beats/min at rest gatran is a useful alternative to warfarin for prevention of stroke or

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


and 90 to 115 beats/min during moderate exercise. embolism in patients with AF. The choice between aspirin and an
A new recommendation as of 2011 is that a resting rate of less than anticoagulant is based on the patient’s risk profile. Factors associated
110 beats/min is a reasonable rate-control target in patients with with the highest risk for thromboembolism are a previous thrombo-
persistent AF and no arrhythmia-related symptoms who have an ejec- embolic event and rheumatic mitral stenosis, and warfarin or dabiga-
tion fraction higher than 40%. However, the guidelines caution that tran is recommended for patients with one of these risk factors. Risk
tachycardia can result in a decline in left ventricular function over factors associated with a moderate risk for thromboembolism are 75
time. years of age or older, hypertension, heart failure, ejection fraction of
Digitalis is much less effective for control of the ventricular rate 35% or lower, and diabetes. Aspirin is recommended if none of these
during exercise than at rest and is indicated for patients with heart risk factors are present, and an anticoagulant is recommended for
failure or left ventricular dysfunction and for sedentary patients. A patients with one or more of these risk factors. In patients with only
combination of digitalis and either a beta blocker or a nondihydro- one of the moderate risk factors, either aspirin or an anticoagulant is
pyridine calcium channel antagonist is appropriate to control the rate reasonable, and the choice should be individualized.
at rest and during exercise. The guidelines recommend that digitalis Rivaroxaban was approved by the Food and Drug Administration
be used as the sole agent for rate control in patients with paroxysmal in 2011 after publication of the updated guidelines dealing with dabi-
AF. Catheter ablation of the AV node should be reserved for patients gatran. It seems appropriate to generalize the recommendations
whose ventricular rate cannot be adequately controlled by drug regarding dabigatran to the factor Xa inhibitor rivaroxaban.
therapy because of either inefficacy or drug intolerance. Another update to the 2006 guidelines addresses combination
therapy with aspirin and clopidogrel. This combination has been
demonstrated to be less effective than warfarin for preventing strokes
Prevention of Thromboembolism but more effective than aspirin by itself. The guidelines now recom-
(Table 38G-2) mend that aspirin plus clopidogrel be considered for prevention of
The 2006 guidelines recommend antithrombotic therapy with either stroke in patients who cannot tolerate or refuse to take an oral
aspirin or a vitamin K antagonist such as warfarin for all patients with anticoagulant.

TABLE 38G-2 ACC/AHA Recommendations for Prevention of Thromboembolism in Atrial Fibrillation


LEVEL OF
CLASS INDICATION EVIDENCE
Class I Antithrombotic therapy to prevent thromboembolism is recommended for all patients with AF, A
(indicated) except those with lone AF or contraindications
Selection of the antithrombotic agent should be based on the absolute risks for stroke and A
bleeding and the relative risk and benefit in a given patient
For patients without mechanical heart valves at high risk for stroke, chronic oral anticoagulant A
therapy with a vitamin K antagonist is recommended in a dose adjusted to achieve the target
intensity INR of 2.0 to 3.0 unless contraindicated. Factors associated with highest risk for stroke
in patients with AF are previous thromboembolism (stroke, transient ischemic attack, or systemic
embolism) and rheumatic mitral stenosis
Anticoagulation with a vitamin K antagonist is recommended for patients with more than one A
moderate risk factor. Such factors include age 75 years or older, hypertension, heart failure,
impaired left ventricular systolic function (ejection fraction ≤35% or fractional shortening
<25%), and diabetes mellitus
The INR should be determined at least weekly during initiation of therapy and monthly when A
anticoagulation is stable
Dabigatran is a useful alternative to warfarin in patients with AF and risk factors for stroke who do B
not have a prosthetic heart valve or significant valve disease, a creatinine clearance <15 mL/min,
or advanced liver disease
Aspirin, 81 to 325 mg daily, is recommended as an alternative to vitamin K antagonists in low-risk A
patients or in those with contraindications to oral anticoagulation
For patients with AF who have mechanical heart valves, the target intensity of anticoagulation B
should be based on the type of prosthesis while maintaining an INR of at least 2.5
Antithrombotic therapy is recommended for patients with atrial flutter as for those with AF C
Class IIa For primary prevention of thromboembolism in patients with nonvalvular AF who have just one A
(reasonable) of the following validated risk factors, antithrombotic therapy with either aspirin or a vitamin K
antagonist is reasonable based on an assessment of the risk for bleeding complications, ability
to safely sustain adjusted chronic anticoagulation, and the patient’s preferences: age 75 years
or older (especially in women), hypertension, heart failure, impaired left ventricular function, or
diabetes mellitus
For patients with nonvalvular AF who have one or more of the following less well validated risk B
factors, antithrombotic therapy with either aspirin or a vitamin K antagonist is reasonable for
prevention of thromboembolism: age 65 to 74 years, female sex, or coronary artery disease.
The choice of agent should be based on the risk for bleeding complications, ability to sustain
adjusted chronic anticoagulation, and the patient’s preferences
It is reasonable to select antithrombotic therapy by the same criteria irrespective of the pattern B
(i.e., paroxysmal, persistent, or permanent) of AF
In patients with AF who do not have mechanical prosthetic heart valves, it is reasonable to C
interrupt anticoagulation for up to 1 week without substituting heparin for surgical or
diagnostic procedures that carry a risk for bleeding
It is reasonable to reevaluate the need for anticoagulation at regular intervals C

Continued
816

TABLE 38G-2 ACC/AHA Recommendations for Prevention of Thromboembolism in Atrial Fibrillation—cont’d


V
LEVEL OF
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

CLASS INDICATION EVIDENCE


Class IIb In patients 75 years or older at increased risk for bleeding but without frank contraindications C
(may be to oral anticoagulant therapy and in other patients with moderate risk factors for
considered) thromboembolism who are unable to safely tolerate anticoagulation at the standard intensity
of an INR of 2.0 to 3.0, a lower INR target of 2.0 (range, 1.6 to 2.5) may be considered for
prevention of ischemic stroke and systemic embolism
When surgical procedures require interruption of oral anticoagulant therapy for longer than 1 C
week in high-risk patients, unfractionated heparin may be administered or low-molecular-
weight heparin given by subcutaneous injection, although the efficacy of these alternatives in
this situation is uncertain
After percutaneous coronary intervention or revascularization surgery in patients with AF, low- C
dose aspirin (<100 mg/day) and/or clopidogrel (75 mg/day) may be given concurrently with
anticoagulation to prevent myocardial ischemic events, but these strategies have not been
thoroughly evaluated and are associated with an increased risk for bleeding
In patients undergoing percutaneous coronary intervention, anticoagulation may be interrupted to C
prevent bleeding at the site of peripheral arterial puncture, but the vitamin K antagonist should
be resumed as soon as possible after the procedure and the dose adjusted to achieve an INR in
the therapeutic range. Aspirin may be given temporarily during the hiatus, but the maintenance
regimen should then consist of the combination of clopidogrel, 75 mg daily, plus warfarin (INR,
2.0 to 3.0). Clopidogrel should be given for a minimum of 1 month after implantation of a bare-
metal stent, at least 3 months for a sirolimus-eluting stent, at least 6 months for a paclitaxel-
eluting stent, and 12 months or longer in selected patients, following which warfarin may be
continued as monotherapy in the absence of a subsequent coronary event. When warfarin is
given in combination with clopidogrel or low-dose aspirin, the dose intensity must be carefully
regulated
In patients with AF younger than 60 years without heart disease or risk factors for C
thromboembolism (lone AF), the risk for thromboembolism is low without treatment, and the
effectiveness of aspirin for primary prevention of stroke relative to the risk for bleeding has not
been established
In patients with AF who sustain an ischemic stroke or systemic embolism during treatment with C
low-intensity anticoagulation (INR, 2.0 to 3.0), rather than add an antiplatelet agent, it may be
reasonable to raise the intensity of the anticoagulation to a maximum target INR of 3.0 to 3.5
Clopidogrel plus aspirin may be considered in patients who cannot tolerate or who refuse an oral B
anticoagulant
Class III (not Long-term anticoagulation with a vitamin K antagonist is not recommended for primary C
indicated) prevention of stroke in patients younger than 60 years without heart disease (lone AF) or any
risk factors for thromboembolism

INR = international normalized ratio.

patients receiving warfarin, heparin should be continued until the


Cardioversion of Atrial Fibrillation INR is 2, and oral anticoagulation with an INR of 2 to 3 should be
(Table 38G-3) continued for 4 or more weeks. In patients treated with dabigatran or
The first-line drugs recommended for cardioversion are flecainide, rivaroxaban, heparin can be discontinued 3 to 4 hours after the first
dofetilide, propafenone, and ibutilide. Amiodarone is considered a oral dose. As with warfarin, anticoagulation therapy should be con-
reasonable option. The guidelines state that digoxin and sotalol may tinued for 4 or more weeks.
be harmful when they are used for cardioversion and recommend
against use of these agents for that purpose.
Direct-current cardioversion is recommended when the ventricular Maintenance of Sinus Rhythm (Table 38G-4)
rate is rapid and does not respond quickly to drug therapy in patients A reasonable outcome of antiarrhythmic drug therapy is infrequent
with myocardial ischemia, hypotension, or heart failure and in recurrences of well-tolerated AF. Initiation of a rhythm-control medi-
patients with Wolff-Parkinson-White syndrome and AF with a very cation is reasonable on an outpatient basis in patients without heart
rapid rate or hemodynamic instability. If AF recurs early after direct- disease when the medication is well tolerated. The updated guide-
current cardioversion, repeated cardioversion is recommended after lines now recommend dronedarone as being a reasonable option in
treatment with an antiarrhythmic drug. patients with paroxysmal AF or after conversion of persistent AF. The
If AF has been present for longer than 48 hours or if the duration guidelines recommend catheter ablation of symptomatic paroxysmal
is unknown, anticoagulation with warfarin and an international nor- AF for patients who have failed treatment with an antiarrhythmic
malized ratio (INR) of 2 to 3 is recommended for 3 or more weeks drug and have little or no left atrial enlargement and normal or mildly
before cardioversion, whether pharmacologic or electrical, and for 4 reduced left ventricular function.
weeks afterward. It is reasonable to presume that dabigatran and
rivaroxaban are suitable alternatives to warfarin.
When cardioversion is performed within 48 hours of the onset of Special Considerations (Table 38G-5)
AF, anticoagulation before and after cardioversion may be based on Postoperative Atrial Fibrillation
the patient’s risk profile. The guidelines recommend prophylactic treatment with an oral beta
In patients with AF lasting longer than 48 hours or in whom the blocker to prevent postoperative AF in patients undergoing cardiac
duration is unknown, an alternative to anticoagulation for 3 or more surgery. Preoperative amiodarone is also considered to be appropri-
weeks before cardioversion is to perform transesophageal echocar- ate prophylactic therapy to prevent postoperative AF. Use of cardio-
diography, to anticoagulate the patient with heparin, to initiate oral version, rhythm-control medications, and antithrombotic medication
anticoagulation, and to proceed with immediate cardioversion if no should be based on the same considerations as in nonsurgical
thrombi are present in the left atrium or left atrial appendage. In patients.
817

TABLE 38G-3 ACC/AHA Recommendations for Cardioversion of Atrial Fibrillation


38
CLASS INDICATION LEVEL OF EVIDENCE

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


Pharmacologic Cardioversion
Class I Administration of flecainide, dofetilide, propafenone, or ibutilide is recommended for pharmacologic A
(indicated) cardioversion of AF
Class IIa Administration of amiodarone is a reasonable option for pharmacologic cardioversion of AF A
(reasonable) A single oral bolus dose of propafenone or flecainide (“pill-in-the-pocket” approach) can be administered C
to terminate persistent AF outside the hospital once treatment has proved safe in the hospital for
selected patients without sinus or AV node dysfunction, bundle branch block, prolongation of the QT
interval, Brugada syndrome, or structural heart disease. Before antiarrhythmic medication is initiated,
a beta blocker or nondihydropyridine calcium channel antagonist should be given to prevent rapid AV
conduction in the event that atrial flutter occurs
Administration of amiodarone can be beneficial on an outpatient basis in patients with paroxysmal or C
persistent AF when rapid restoration of sinus rhythm is not deemed necessary
Class IIb (may be Administration of quinidine or procainamide might be considered for pharmacologic cardioversion of AF, C
considered) but the usefulness of these agents is not well established
Class III (not Digoxin and sotalol may be harmful when used for pharmacologic cardioversion of AF and are not A
indicated) recommended
Quinidine, procainamide, disopyramide, and dofetilide should not be started out of the hospital for B
conversion of AF to sinus rhythm
Direct-Current Cardioversion
Class I When a rapid ventricular response to pharmacologic measures does not occur in patients with AF and C
(indicated) ongoing myocardial ischemia, symptomatic hypotension, angina, or heart failure, immediate R wave–
synchronized direct-current cardioversion is recommended
Immediate direct-current cardioversion is recommended for patients with AF involving preexcitation when B
very rapid tachycardia or hemodynamic instability occurs
Cardioversion is recommended in patients without hemodynamic instability when symptoms of AF are C
unacceptable to the patient. In case of early relapse of AF after cardioversion, repeated attempts at
direct-current cardioversion may be made after administration of antiarrhythmic medication
Class IIa Direct-current cardioversion can be useful to restore sinus rhythm as part of a long-term management B
(reasonable) strategy for patients with AF
The patient’s preference is a reasonable consideration in the selection of infrequently repeated C
cardioversions for the management of symptomatic or recurrent AF
Class III (not Frequent repetition of direct-current cardioversion is not recommended for patients who have relatively C
indicated) short periods of sinus rhythm between relapses of AF after multiple cardioversion procedures despite
prophylactic antiarrhythmic drug therapy
Electrical cardioversion is contraindicated in patients with digitalis toxicity or hypokalemia C
Pharmacologic Enhancement of Direct-Current Cardioversion
Class IIa Pretreatment with amiodarone, flecainide, ibutilide, propafenone, or sotalol can be useful to enhance the B
(reasonable) success of direct-current cardioversion and to prevent recurrent AF
In patients in whom AF recurs after successful cardioversion, it can be useful to repeat the procedure C
after prophylactic administration of antiarrhythmic medication
Class IIb (may be For patients with persistent AF, administration of beta blockers, disopyramide, diltiazem, dofetilide, C
considered)) procainamide, or verapamil may be considered, although the efficacy of these agents in enhancing the
success of direct-current cardioversion or in preventing early recurrence of AF is uncertain
Out-of-hospital initiation of antiarrhythmic medications may be considered in patients without heart C
disease to enhance the success of cardioversion of AF
Out-of-hospital administration of antiarrhythmic medications may be considered to enhance the success C
of cardioversion of AF in patients with certain forms of heart disease once the safety of the drug has
been verified for the patient
Prevention of Thromboembolism in Patients with Atrial Fibrillation Undergoing Cardioversion
Class I For patients with AF lasting 48 hours or longer or when the duration of AF is unknown, anticoagulation B
(indicated) (INR of 2.0 to 3.0) is recommended for at least 3 weeks before and 4 weeks after cardioversion,
regardless of the method (electrical or pharmacologic) used to restore sinus rhythm
For patients with AF lasting longer than 48 hours who require immediate cardioversion because of C
hemodynamic instability, heparin should be administered concurrently (unless contraindicated) via an
initial intravenous bolus injection, followed by a continuous infusion in a dose adjusted to prolong the
activated partial thromboplastin time to 1.5 to 2 times the reference control value. Thereafter, oral
anticoagulation (INR of 2.0 to 3.0) should be provided for at least 4 weeks, as for patients undergoing
elective cardioversion. Limited data support subcutaneous administration of low-molecular-weight
heparin for this indication
For patients with AF of less than a 48-hour duration associated with hemodynamic instability (angina C
pectoris, myocardial infarction, shock, or pulmonary edema), cardioversion should be performed
immediately, without delay for initiation of anticoagulation

Continued
818

TABLE 38G-3 ACC/AHA Recommendations for Cardioversion of Atrial Fibrillation—cont’d


V
CLASS INDICATION LEVEL OF EVIDENCE
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

Class IIa During the 48 hours after the onset of AF, the need for anticoagulation before and after cardioversion C
(reasonable) may be based on the patient’s risk for thromboembolism
As an alternative to anticoagulation before cardioversion of AF, it is reasonable to perform B
transesophageal echocardiography to search for a thrombus in the left atrium or left atrial appendage
a. For patients with no identifiable thrombus, cardioversion is reasonable immediately after B
anticoagulation with unfractionated heparin (e.g., initiated by intravenous bolus injection and an
infusion continued at a dose adjusted to prolong the activated partial thromboplastin time to 1.5
to 2 times the control value until oral anticoagulation has been established with an oral vitamin K
antagonist [e.g., warfarin] as evidenced by an INR ≥2.0)
Thereafter, continuation of oral anticoagulation (INR of 2.0 to 3.0) is reasonable for a total B
anticoagulation period of at least 4 weeks, as for patients undergoing elective cardioversion
Limited data are available to support the subcutaneous administration of a low-molecular-weight C
heparin for this indication
b. For patients in whom a thrombus is identified by transesophageal echocardiography, oral C
anticoagulation (INR of 2.0 to 3.0) is reasonable for at least 3 weeks before and 4 weeks after
restoration of sinus rhythm, and a longer period of anticoagulation may be appropriate even after
apparently successful cardioversion because the risk for thromboembolism often remains elevated in
such cases
For patients with atrial flutter undergoing cardioversion, anticoagulation can be beneficial according C
to the recommendations as for patients with AF

TABLE 38G-4 ACC/AHA Recommendations for Maintenance of Sinus Rhythm in Patients with Atrial Fibrillation
CLASS INDICATION LEVEL OF EVIDENCE
Class I (indicated) Before initiation of antiarrhythmic drug therapy, treatment of precipitating or reversible C
causes of AF is recommended
Catheter ablation by an experienced operator is useful in selected patients with A
symptomatic paroxysmal AF who have failed treatment with an antiarrhythmic drug
and have a normal or mildly dilated left atrium and normal or mildly reduced left
ventricular function
Class IIa (reasonable) Pharmacologic therapy can be useful in patients with AF to maintain sinus rhythm and C
to prevent tachycardia-induced cardiomyopathy
Infrequent, well-tolerated recurrence of AF is reasonable as a successful outcome of C
antiarrhythmic drug therapy
Outpatient initiation of antiarrhythmic drug therapy is reasonable in patients with AF C
who have no associated heart disease when the agent is well tolerated
In patients with lone AF without structural heart disease, initiation of propafenone or B
flecainide can be beneficial on an outpatient basis in patients with paroxysmal AF
who are in sinus rhythm at the time of drug initiation
Sotalol can be beneficial in outpatients in sinus rhythm with little or no heart disease C
who are prone to paroxysmal AF if the baseline uncorrected QT interval is shorter
than 460 msec, serum electrolyte values are normal, and risk factors associated with
class III drug–related proarrhythmia are not present
Catheter ablation is a reasonable option for the treatment of symptomatic persistent AF A
Class IIb (may be Catheter ablation may be reasonable for patients with symptomatic paroxysmal AF and A
considered) significant left atrial dilation or significant left ventricular dysfunction
Class III (not Antiarrhythmic therapy with a particular drug is not recommended for maintenance A
indicated) of sinus rhythm in patients with AF who have well-defined risk factors for
proarrhythmia with that agent
Pharmacologic therapy is not recommended for maintenance of sinus rhythm in C
patients with advanced sinus node disease or AV node dysfunction unless they have
a functioning electronic cardiac pacemaker

Acute Myocardial Infarction Atrial Fibrillation in Wolff-Parkinson-White Syndrome


Electrical cardioversion is recommended for patients with hemody- Catheter ablation of the accessory pathway is recommended for
namic compromise or ongoing ischemia or when adequate rate patients with symptomatic AF and Wolff-Parkinson-White syndrome.
control cannot be achieved with drug therapy. The guidelines recom- Immediate electrical cardioversion is recommended for those with
mend intravenous amiodarone or digitalis to slow the ventricular rate AF and a rapid ventricular rate and hemodynamic instability. If
in patients and to improve left ventricular function in those with acute the patient is hemodynamically stable, intravenous procainamide or
myocardial infarction. In patients without left ventricular dysfunc- ibutilide is recommended for pharmacologic conversion of AF. Intra-
tion, bronchospasm, or AV block, an intravenous beta blocker or venous digitalis and nondihydropyridine calcium channel antago-
nondihydropyridine calcium channel antagonist is recommended for nists should be avoided in patients with ventricular preexcitation
rate control. during AF.
819

TABLE 38G-5 ACC/AHA Recommendations for Special Considerations in Atrial Fibrillation


38
CLASS INDICATION LEVEL OF EVIDENCE

Atrial Fibrillation: Clinical Features, Mechanisms, and Management


Postoperative Atrial Fibrillation
Class I Unless contraindicated, treatment with an oral beta blocker to prevent postoperative AF is A
(indicated) recommended for patients undergoing cardiac surgery
Administration of AV nodal blocking agents is recommended to achieve rate control in patients in B
whom AF develops postoperatively
Class IIa Preoperative administration of amiodarone reduces the incidence of AF in patients undergoing cardiac A
(reasonable) surgery and represents appropriate prophylactic therapy for patients at high risk for postoperative AF
It is reasonable to restore sinus rhythm by pharmacologic cardioversion with ibutilide or direct-current B
cardioversion in patients in whom AF develops postoperatively, as advised for nonsurgical patients
It is reasonable to administer antiarrhythmic medications in an attempt to maintain sinus rhythm in B
patients with recurrent or refractory postoperative AF, as recommended for other patients in whom
AF develops
It is reasonable to administer antithrombotic medication in patients in whom AF develops B
postoperatively, as recommended for nonsurgical patients
Class IIb (may be Prophylactic administration of sotalol may be considered for patients at risk for the development of AF B
considered) after cardiac surgery
Acute Myocardial Infarction
Class I Direct-current cardioversion is recommended for patients with severe hemodynamic compromise or C
(indicated) intractable ischemia or when adequate rate control cannot be achieved with pharmacologic agents
in patients with acute myocardial infarction and AF
Intravenous administration of amiodarone is recommended to slow a rapid ventricular response to AF C
and to improve left ventricular function in patients with acute myocardial infarction
Intravenous beta blockers and nondihydropyridine calcium channel antagonists are recommended C
to slow a rapid ventricular response to AF in patients with acute myocardial infarction who do not
display clinical left ventricular dysfunction, bronchospasm, or AV block
For patients with AF and acute myocardial infarction, administration of unfractionated heparin by C
either continuous intravenous infusion or intermittent subcutaneous injection is recommended in
a dose sufficient to prolong the activated partial thromboplastin time to 1.5 to 2 times the control
value, unless contraindications to anticoagulation exist
Class IIa Intravenous administration of digitalis is reasonable to slow a rapid ventricular response and to improve C
(reasonable) left ventricular function in patients with acute myocardial infarction and AF associated with severe
left ventricular dysfunction and heart failure
Class III (not Administration of class IC antiarrhythmic drugs is not recommended in patients with AF in the setting C
indicated) of acute myocardial infarction
Management of Atrial Fibrillation Associated with Wolff-Parkinson-White (WPW) Preexcitation Syndrome
Class I Catheter ablation of the accessory pathway is recommended for symptomatic patients with AF who B
(indicated) have WPW syndrome, particularly those with syncope secondary to a rapid heart rate or those with
a short bypass tract refractory period
Immediate direct-current cardioversion is recommended to prevent ventricular fibrillation in patients B
with a short anterograde bypass tract refractory period in whom AF occurs with a rapid ventricular
response associated with hemodynamic instability
Intravenous procainamide or ibutilide is recommended to restore sinus rhythm in patients with WPW C
syndrome in whom AF occurs without hemodynamic instability in association with a wide QRS
complex on the electrocardiogram (≥120-msec duration) or with a rapid preexcited ventricular
response
Class IIa Intravenous flecainide or direct-current cardioversion is reasonable when very rapid ventricular rates B
(reasonable) occur in patients with AF involving conduction over an accessory pathway
Class IIb (may be It may be reasonable to administer intravenous quinidine, procainamide, disopyramide, ibutilide, or B
considered) amiodarone to hemodynamically stable patients with AF involving conduction over an accessory
pathway
Class III (not Intravenous administration of digitalis glycosides or nondihydropyridine calcium channel antagonists B
indicated) is not recommended in patients with WPW syndrome who have preexcited ventricular activation
during AF
Hyperthyroidism
Class I Administration of a beta blocker is recommended to control the rate of ventricular response in patients B
(indicated) with AF complicating thyrotoxicosis, unless contraindicated
In circumstances in which a beta blocker cannot be used, administration of a nondihydropyridine B
calcium channel antagonist (diltiazem or verapamil) is recommended to control the ventricular rate
in patients with AF and thyrotoxicosis
In patients with AF associated with thyrotoxicosis, oral anticoagulation (INR of 2.0 to 3.0) is C
recommended to prevent thromboembolism, as recommended for AF patients with other risk
factors for stroke
Once a euthyroid state is restored, recommendations for antithrombotic prophylaxis are the same as C
for patients without hyperthyroidism

Continued
820

TABLE 38G-5 ACC/AHA Recommendations for Special Considerations in Atrial Fibrillation—cont’d


V
CLASS INDICATION LEVEL OF EVIDENCE
ARRHYTHMIAS, SUDDEN DEATH, AND SYNCOPE

Management of Atrial Fibrillation During Pregnancy


Class I Digoxin, a beta blocker, or a nondihydropyridine calcium channel antagonist is recommended to C
(indicated) control the rate of ventricular response in pregnant patients with AF
Direct-current cardioversion is recommended in pregnant patients who become hemodynamically C
unstable because of AF
Protection against thromboembolism is recommended throughout pregnancy for all patients with C
AF (except those with lone AF and/or low thromboembolic risk). Therapy (anticoagulant or aspirin)
should be chosen according to the stage of pregnancy
Class IIb (may be Administration of heparin may be considered during the first trimester and last month of pregnancy B
considered) for patients with AF and risk factors for thromboembolism. Unfractionated heparin may be
administered either by continuous intravenous infusion in a dose sufficient to prolong the activated
partial thromboplastin time to 1.5 to 2 times the control value or by intermittent subcutaneous
injection in a dose of 10,000 to 20,000 units every 12 hours, adjusted to prolong the midinterval (6
hours after injection) activated partial thromboplastin time to 1.5 times control
Despite the limited data available, subcutaneous administration of low-molecular-weight heparin may C
be considered during the first trimester and last month of pregnancy for patients with AF and risk
factors for thromboembolism
Administration of an oral anticoagulant may be considered during the second trimester for pregnant C
patients with AF and high thromboembolic risk
Administration of quinidine or procainamide may be considered to achieve pharmacologic C
cardioversion in hemodynamically stable patients in whom AF develops during pregnancy
Management of Atrial Fibrillation in Patients with Hypertrophic Cardiomyopathy (HCM)
Class I Oral anticoagulation (INR of 2.0 to 3.0) is recommended in patients with HCM in whom AF develops, B
(indicated) as for other patients at high risk for thromboembolism
Class IIa (may be Antiarrhythmic medications can be useful to prevent recurrent AF in patients with HCM. The C
considered) data available are insufficient to recommend one agent over another in this situation, but (1)
disopyramide combined with a beta blocker or nondihydropyridine calcium channel antagonist or (2)
amiodarone alone is generally preferred
Management of Atrial Fibrillation in Patients with Pulmonary Disease
Class I Correction of hypoxemia and acidosis is the recommended primary therapeutic measure for patients in C
(indicated) whom AF develops during an acute pulmonary illness or exacerbation of chronic pulmonary disease
A nondihydropyridine calcium channel antagonist (diltiazem or verapamil) is recommended to control C
the ventricular rate in patients with obstructive pulmonary disease in whom AF develops
Direct-current cardioversion should be attempted in patients with pulmonary disease who become C
hemodynamically unstable as a consequence of AF
Class III (not Theophylline and beta-adrenergic agonist agents are not recommended for patients with C
indicated) bronchospastic lung disease in whom AF develops
Beta blockers, sotalol, propafenone, and adenosine are not recommended in patients with obstructive C
lung disease in whom AF develops

Hyperthyroidism Hypertrophic Cardiomyopathy


The guidelines recommend a beta blocker as first-line therapy for rate The guidelines point out that data are not adequate to determine the
control in patients with AF and thyrotoxicosis. If a beta blocker cannot best rhythm-control medication to use for AF in the setting of hyper-
be used, verapamil or diltiazem should be used for rate control. trophic cardiomyopathy. The preferred therapy is either disopyra-
Recommendations for therapy to prevent thromboembolic complica- mide plus a beta blocker, verapamil, or diltiazem for rate control or
tions are the same as for patients without hyperthyroidism. amiodarone by itself.

Atrial Fibrillation during Pregnancy


The guidelines recommend digoxin, a beta blocker, or a nondihydro- Pulmonary Disease
pyridine calcium channel antagonist for rate control of AF during The primary therapy for AF in the setting of an acute pulmonary
pregnancy. Direct-current cardioversion is recommended in hemody- illness or exacerbation of chronic pulmonary disease should be cor-
namically unstable patients. rection of hypoxemia and acidosis. Verapamil or diltiazem is recom-
Except in patients with a low-risk profile, either aspirin or an anti- mended for rate control in patients with obstructive pulmonary
coagulant is recommended for prevention of thromboembolic com- disease. Theophylline and beta-adrenergic agonists are not recom-
plications, depending on the stage of pregnancy (see Chapter 82). mended in patients with bronchospastic disease, and beta blockers,
Unfractionated or low-molecular-weight heparin can be considered sotalol, propafenone, and adenosine are not recommended in
during the first trimester and last month of pregnancy in patients with patients with obstructive lung disease.
risk factors for thromboembolism, and an oral anticoagulant can be
considered during the second trimester in patients at high risk for
thromboembolism. References
When AF occurs during pregnancy, quinidine or procainamide can 1. Fuster V, Ryden LE, Cannom DS, et al: 2011 ACCF/AHA/HRS focused updates incorporated
into the ACC/AHA/ESC 2006 guidelines for the management of patients with atrial fibrillation:
be considered for pharmacologic cardioversion in hemodynamically A report of the American College of Cardiology Foundation/American Heart Association task
stable patients. force on practice guidelines. Circulation 123:e269, 2011.

Potrebbero piacerti anche