Canadian Cardiovascular Society
Guideline Toolbar

9. Arrhythmia Management

9.1 Acute management of AF

The acute management of AF is centred on the following domains:

1. Determination if AF is the primary concern (“primary AF”) or secondary to another acute medical illness (“secondary AF”). AF in the setting of critical illness has been associated with an increased risk of death (see section 11.5).[505],[506] Unfortunately, there is a paucity of high-quality evidence on whether or how to treat AF patients in the setting of critical illness,[507],[508] and there is a wide variety of reported approaches to AF management in this setting.[509] In the ED setting, results of a retrospective study suggested that rate and rhythm control efforts in patients with AF secondary to acute medical illness (predominantly sepsis and acute HF) might be associated with higher rates of adverse events.[510]

2. Determination of hemodynamic stability, defined as AF causing hypotension, ACS, or pulmonary edema. Acute unstable AF should be treated with synchronized direct current cardioversion (DCCV), however, instability solely due to AF is rare, therefore, an underlying precipitant should also be aggressively sought and managed.[511]

3. Determination of an arrhythmia management strategy, defined as rate vs rhythm control. In patients with established AF multiple RCTs have shown no significant difference in cardiovascular outcomes between patients treated with a strategy with rate control vs rhythm control.[512][514] In patients with newly diagnosed AF (ie, within a year) an initial strategy of rhythm control has been associated with reduced cardiovascular death and reduced rates of stroke.[515]

4. Determination of the need for hospitalization. Most patients with AF can be safely discharged home after acute management. However, hospitalization might be required for highly symptomatic patients with AF in association with acute medical illness or complex medical conditions, in highly symptomatic patients in whom adequate rate control cannot be achieved, or in those who require monitoring or ancillary investigations not readily available in the outpatient setting.

5. Determination of the need for OAC. There is evidence that OAC prescription in the ED results in improved longterm use.[364],[516],[517] As such, it is of paramount importance that OAC be initiated as soon as time allows in patients who undergo cardioversion for new-onset AF (see section 8.4.1), as well as in patients at risk of stroke (see the “CCS algorithm” in Fig. 8) whether or not attempts at rhythm control are made.

6. Early follow-up. Patients discharged from the ED with AF benefit from early follow-up. Ideally this should occur within a week of discharge, because early follow-up has been associated with lower rates of readmission and death.[518] In addition, cardiology assessment within 3 months of hospital discharge for new-onset AF has been associated with lower rates of death, stroke, and major bleeding.[519]

A general overview of rate and rhythm management of AF is provided in Figure 16, and the approach to the management of AF in the acute care setting is provided in Figure 17.

Recommendation

70. We recommend that the management of patients who present with recent-onset AF due to a reversible or secondary cause should be directed at the primary illness (Strong Recommendation; Low-Quality Evidence).

Values and Preferences

AF in the acute care environment can be secondary to a primary cardiac pathology or can occur secondary to a specific precipitating event, such as infection, surgery, or thyroid disease.

71. We recommend immediate electrical cardioversion for patients whose recent-onset AF is the direct cause of instability with hypotension, ACS, or pulmonary edema (Strong Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a high value on immediately addressing instability by attempting cardioversion and a lower value on reducing the risk of cardioversion-associated stroke with a period of anticoagulation before cardioversion.

Practical Tip

Therapeutic anticoagulation should be initiated as soon as possible, ideally prior to cardioversion if time allows.

72. We suggest that a rhythm control strategy be considered for most stable patients with recent-onset AF (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

In patients with established AF multiple RCTs have shown no significant difference in cardiovascular outcomes between patients treated with a strategy with rate control vs rhythm control, recognizing that most of these trials did not specifically address recentonset AF. In patients with newly diagnosed AF (ie, within a year) an initial strategy of rhythm control has been associated with reduced cardiovascular death and reduced rates of stroke.

73. In patients with AF and manifest pre-excitation we recommend against acute rate control (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

See section 11.8 for the management of patients with AF and pre-excitation.

74. We recommend that patients who present with AF in the acute care setting have their need for long-term antithrombotic therapy be determined using the CCS Algorithm (CHADS-65) (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

See section 8.4.1 for the recommendations regarding OAC in the context of cardioversion.

9.1.1 Acute rate control

In the setting of recent onset AF, the rate control agent and the formulation chosen will be influenced by clinical circumstance (eg, the presence of HF or hypotension) and patient comorbidities (eg, known LV dysfunction, reactive airways disease, hypotension, history of MI, or angina; Supplemental Table S10). Options include oral or I.V. β-blockers, oral or I.V. nondihydropyridine calcium channel blockers (ND-CCBs), I.V. digoxin, and I.V. amiodarone (recognizing that the latter is also a rhythm control agent). I.V. rate control agents might be initially considered, however if the patient is hemodynamically stable oral agents might be preferred. If an I.V. agent is used as the initial therapy, it is important to coadminister an oral rate control agent as soon as possible to maintain rate control/avoid rebound tachycardia as the I.V. formulation wears off.[520] In patients without contraindications, β-blockers and calcium channel blockers (CCBs) are considered first-line agents for rate control. Only two small RCTs totalling 92 patients have compared I.V. β-blockers with CCBs in patients with recent-onset AF (one of which also included AFL), both showed that I.V. diltiazem was more effective at controlling the heart rate (< 100 beats per minute [bpm]) at 20-30 minutes compared with I.V. metoprolol (RR, 1.8; 95% CI, 1.2- 2.6).[521][523] A retrospective study of 110 AF patients showed similar results at 60 minutes, although the success rate with diltiazem in that study (57%) was lower than those reported in the RCTs (90%-95%).[521],[522],[524] In patients with ACS who require acute rate control, β-blockers are the agent of choice. Digoxin or amiodarone might be considered for acute rate control in the setting of decompensated HF, known significant LV systolic dysfunction (defined as LVEF ≤ 40%), or mild hypotension.[7] However, it is important to recall that I.V. formulations of amiodarone can lower BP.[525] Moreover, in this population the selective use of I.V. CCBs (and β-blockers) has been safely and successfully used in several randomized and nonrandomized studies, often with an improvement in BP when the recent-onset AF is rate controlled. Jandali performed a retrospective cohort study on the use of I.V. diltiazem in 162 patients with LV systolic dysfunction (LVEF ≤ 50%; with 52 having an LVEF ≤ 30%), and compared them with 473 patients with preserved LVEF (≥ 50%).[526] There was no difference in the rates of hypotension, intensive care unit (ICU) transfer, or mortality between the patients with LVEF ≥ 50% vs those with LVEF < 50%, or those with LVEF ≥ 30% vs those with LVEF < 30%. Hirschy et al. performed a retrospective cohort study on the use of I.V. metoprolol (14 patients) and I.V. diltiazem (34 patients) in patients with known LV systolic dysfunction (mean LVEF 23% [15-35] vs 25% [15-30], respectively). Successful rate control within 30 minutes occurred in 62% of the metoprolol group and 50% of the diltiazem group (P ¼ 0.49), with no difference in complications or worsening HF.[527] Goldenberg et al. performed a small double-blind RCT of 37 AF patients with reduced ejection fraction (EF; LVEF 36% ± 14%) and symptomatic HF (New York Heart Association [NYHA] class III [62%] and IV [38%]), in whom the cautious use of I.V. diltiazem resulted in therapeutic response (97%) with self-limited hypotension in 11%, and no patients experiencing worsening of HF.[528] Although these studies have shown that the use of I.V. CCBs for rate control in highly selected patients can be safe, caution must be used because of the risk of precipitating cardiac decompensation.

Recommendation

75. We recommend that either β-blockers or ND-CCBs (diltiazem or verapamil) be first-line agents for AF rate control in patients without significant LV dysfunction (eg, patients with an LVEF > 40%) (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

Use caution when administering I.V. formulations of β-blockers or ND-CCBs (verapamil and diltiazem) because of the risk of precipitating hypotension.

Practical Tip

The selection of a β-blocker or NDCCB for rate control of AF should be on the basis of patient comorbidities, contraindications, and side effect profile.

Practical Tip

Oral formulations should be introduced as soon as possible because of the need for ongoing control of ventricular rate.

76. We recommend evidence-based β-blockers (bisoprolol, carvedilol, metoprolol) be first-line agents for rate control of hemodynamically stable AF in the acute care setting in patients with significant LV dysfunction (LVEF ≤ 40%) (Strong Recommendation; Moderate-Quality Evidence).

77. We suggest I.V. amiodarone or I.V. digoxin be considered for acute rate control in patients with significant LV dysfunction (LVEF ≤ 40%), decompensated HF, or hypotension, when immediate electrical cardioversion is not indicated (Weak Recommendation; Moderate-Quality Evidence).

Practical Tip

Use caution when administering I.V. amiodarone for rate control because of the possibility of hypotension and/or conversion to sinus rhythm, and the subsequent risk of stroke in patients who are not adequately anticoagulated.

9.1.1.1 Acute rate control targets

There have been no RCTs that specifically examined rate control targets in the acute care setting,[529] nor in patients with paroxysmal AF.[530]

Recommendation

78. We recommend titrating rate-controlling agents to achieve a heart rate target of ≤ 100 bpm at rest for patients who present with a primary diagnosis of AF in the acute care setting (Strong Recommendation; Low-Quality Evidence).

Practical Tip

There is no evidence to support a specific heart rate target in acutely ill patients with AF secondary to a reversible or secondary cause. Treatment targets should be individualized in this patient population after consideration of the risk/benefits of pharmacological rate control.

9.1.2 Acute rhythm control

For stable patients with recent-onset AF who are eligible for cardioversion, the choice to pursue sinus rhythm restoration should be made on the basis of patient symptoms and goals of care, recognizing that early rhythm control has been associated with a lower risk of stroke and cardiovascular death.[515] Because cardioversion increases the risk of systemic embolism, it is important to start appropriate anticoagulation as soon as time allows for all patients (see section 8.4.1).[339] For patients with recent-onset AF who are eligible for cardioversion, rhythm control is preferred and can be established via either pharmacological or electrical cardioversion. In general, electrical cardioversion is more effective than pharmacological cardioversion, especially for more prolonged AF episode durations.[363],[531][534] Pharmacological cardioversion has the advantage of being immediately feasible in a nonfasting patient, as well as avoiding the delays and risks associated with procedural sedation. However, most pharmacological agents have cautions or contraindications that limiting their use in patients with significant cardiac comorbidities, and their use requires a monitored bed, access to a crash cart, and a dedicated nurse to monitor for potential complications.

Recommendation

79. We recommend that synchronized direct current or pharmacologic cardioversion may be used for sinus rhythm restoration in hemodynamically stable patients with recent-onset AF (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

In treatment environments in which procedural sedation is readily available, DCCV might be the preferred initial means to restore sinus rhythm, because it is more than 90% effective in the acute care environment and reduces ED length of stay.

Practical Tip

A strategy of pharmacological conversion followed by DCCV (if necessary) might be preferred in environments in which procedural sedation is not readily available, because pharmacological conversion might avert the need for DCCV in approximately half of the treated patients.

9.1.2.1 Pharmacologic cardioversion

Antiarrhythmic medication selection is typically dictated by the patient’s comorbidities as well as physician preference. Characteristics, indications, contraindications, and monitoring details of antiarrhythmic medications used for acute pharmacological cardioversion are presented in Supplemental Table S11.

Recommendation

80. We recommend that the choice of antiarrhythmic drug used for acute pharmacological cardioversion be defined according to patient characteristics (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

The choice of medication in patients without any contraindications will depend on physician experience, duration of AF, and considerations unique to the practice setting (See Supplemental Table S11).

Practical Tip

All patients require monitoring after cardioversion, the length of which is dependent on the method of conversion. A general guideline is to observe patients for a duration of time that is equal to half of the medication’s therapeutic half-life.

Practical Tip

If the patient’s history is unknown, electrical cardioversion should be used in preference to pharmacological cardioversion.

9.1.2.1.1 Procainamide

Procainamide, a class Ia agent, is the most common I.V. medication used for cardioversion of recent-onset AF in the Canadian ED setting.[535] Although procainamide can be administered as a 1-g bolus over 30 minutes followed by an infusion of 2 mg/min or a dose of 15-18 mg/kg administered over 60 minutes, most clinical evidence (including safety outcomes) was derived on the basis of a single infusion of 1 g over 60 minutes.[531],[536][538] Procainamide is more effective for the conversion of recent-onset AF (50%-60% conversion) than for AFL (30% conversion).[531],[536][538] The most common side effect is hypotension (approximately 5%), although premature ventricular contractions, runs of ventricular tachycardia and QRS widening might occur.[531],[536],[537] This medication, like all drugs with class I (Na+ channel-blocking) action, should be avoided in patients with Brugada syndrome.[539]

9.1.2.1.2 Ibutilide

Ibutilide is an I.V. class III agent that has been shown to effectively terminate AFL (50%-75%) and AF (30%-50%), with cardioversion typically occurring within 30-60 minutes.[540][546] However, widespread clinical uptake has been limited by a significant risk of torsades de pointes (TdP) and ventricular tachycardia (most frequently nonsustained), each of which occurs in approximately 2%-3% of patients.[540][548] Consequently, ibutilide should not be used in patients with a prolonged QTc (> 440 ms), a history of HF (typically defined as clinically symptomatic or NYHA classification > II), or reduced EF, signs of an ACS, and/or low serum potassium or magnesium levels.[540][545],[547] Periprocedural I.V. magnesium (typically given pre- and post treatment) appears to improve ibutilide cardioversion rates, with higher doses (eg, 4 g total) more effective than lower doses (1-3 g total).[549],[550] Patients must be observed with continuous ECG monitoring for a minimum of 4 hours after ibutilide administration.

9.1.2.1.3 Vernakalant

Vernakalant is an atrial-selective antiarrhythmic approved for conversion of AF. In patients treated within 48 hours of AF onset, randomized trials report a conversion rate at 90 minutes ranging from 52% to 69%, which is not significantly better than other active agents (combined comparator of ibutilide and amiodarone).[544],[545],[551][553] However, the median time to cardioversion of 10-12 minutes is shorter than the next fastest pharmacological agent (ibutilide, median time to conversion 26 minutes). The major side effects are hypotension and bradycardia after cardioversion.[551],[554],[555] Transient but fairly common side effects include dysgeusia, paresthesia, and nausea.[544],[551],[554],[555] Vernakalant is not effective for the conversion of AFL; and should be avoided in patients with hypotension, severe HF (NYHA classification III/IV), bradycardia, recent ACS, or severe aortic stenosis.[552],[555],[556]

9.1.2.1.4 Amiodarone

With the exception of patients with structural heart disease, amiodarone is not recommended for acute rhythm control because of a delay in conversion (approximately 8 hours).[525],[532],[557] The most common adverse drug reactions with I.V. administration are phlebitis, hypotension, and bradycardia.[525],[532] Although there is potential for prolongation of the QT interval, the incidence of TdP is rare.[532],[557]

9.1.2.1.5 Flecainide and propafenone

I.V. flecainide and propafenone are superior to placebo but are not currently available in Canada for acute care cardioversion.[525],[558] The oral formulations, however, have similar, if slightly delayed, efficacy as their I.V. counterparts.[558],[559] Three hours after administration of a single dose of oral flecainide, between 57% and 68% of patients will convert.[532] Success rates with oral propafenone are similar.[532],[559] Although the time to cardioversion (approximately 2-6 hours) is longer than with I.V. formulations, the major clinical benefit is that patients are able to treat their AF episodes at home (“pill-in-the-pocket”), which reduces the need to visit the ED for recurrences. A key caveat to this approach is that the first treatment attempt must be administered in a monitored environment, to verify efficacy and exclude treatment-related adverse reactions.[557],[560][563] A β-blocker or ND-CCB should be given ≥ 30 minutes before administration of a class Ic antiarrhythmic to prevent the risk of 1:1 AV conduction during AFL. One study suggests that rare adverse events can occur even after successful use in a monitored environment[563]; therefore, clear instructions must be given to these patients about when to seek emergency care (Supplemental Table S12). It is important to note that flecainide and propafenone should not be used in patients with structural heart disease, including a history of ischemic heart disease.

9.1.2.2 Electrical cardioversion

In patients with hemodynamically stable recent-onset AF in whom sinus rhythm restoration is desired, there is a wealth of observational data supporting the safety and efficacy (approximately 90%) of direct-current cardioversion (DCCV).[363],[531],[533],[535],[564][567] Since the late 1990s patient sedation and cardioversion have typically been performed by emergency physicians in the Canadian ED setting.[531],[535],[565] Adverse events attributable to DCCV such as bradyarrhythmia, acute HF, and skin burns are rare.[533],[566],[567] The time to patient discharge using DCCV is shorter than when using pharmacological cardioversion.[534],[568] Importantly, AF patients who receive DCCV in the ED rate their care as more effective compared with those who receive only rate control, however, their QOL scores at 30 days were not different than those treated with only rate control.[569] Biphasic shocks are preferred over monophasic because less energy is required.[570] Pad placement (anterolateral vs anteroposterior) does not seem to influence cardioversion efficacy.[538],[571] In obese patients, using paddles and applying force might improve success rates with DCCV over adhesive pads.[572],[573] Pretreatment with antiarrhythmic drugs (eg, ibutilide and amiodarone) has been shown to improve the effectiveness of DCCV.[363],[574]

Recommendation

81. We recommend at least a 150-J biphasic waveform as the initial energy setting for DCCV (Strong Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a high value on the avoidance of repeated shocks after ineffective attempts at low-energy cardioversion.

Practical Tip

Electrical cardioversion should ideally be performed with one trained operator managing the sedation and airway and a second trained operator managing the synchronized DCCV. Atropine and pacing capability must be immediately available in case of prolonged sinus pause after cardioversion.

82. We suggest antiarrhythmic drug therapy be considered to enhance the efficacy of electrical cardioversion and the maintenance of sinus rhythm, particularly in patients with persistent and long-standing persistent AF (Weak Recommendation; Low-Quality Evidence).

83. We suggest that the use of antiarrhythmic drug therapy after sinus rhythm restoration be on the basis of the estimated probability of AF recurrence (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a high value on minimizing the risk of infrequent but serious side effects associated with long-term antiarrhythmic drugs. A high value is also placed on the appropriate use of speciality care to make patient-specific decisions to minimize these risks.

9.2 Long-term rate control

9.2.1 Agents

Pharmacotherapy for long-term AF rate control revolves around agents with negative dromotropic properties such as β-blockers and ND-CCBs (verapamil and diltiazem). The choice of a specific rate-controlling regimen should be on the basis of patient’s characteristics and the drug’s efficacy/side effect profile (Supplemental Table S10; Fig. 18).[575] In patients without significant LV dysfunction (LVEF > 40%), β-blockers and ND-CCBs are first-line options. There are no randomized long-term data to support choosing a β-blocker over an ND-CCB. Several retrospective studies of AF patients have shown conflicting results when rates of hospital admission after using β-blockers vs CCBs were compared: one showed no difference whereas another showed that use of CCBs was associated with a higher rate of hospitalization compared with use of β-blockers.[520],[576] In the longer-term, β-blockers might be more effective at slowing ventricular rates at rest and during exercise, however, their use is associated with a higher risk of adverse effects, notably fatigue and exercise intolerance.[577][580] Moreover, there is emerging evidence suggesting that CCBs might have favourable dose-response characteristics for AF rate control vs β-blockers, such that they might be preferred in patients with a preserved LVEF and without another indication for a β-blocker.[522],[580][582] Specific patient characteristics might favour the use of one pharmacological class (eg, ND-CCBs with hypertension or reactive airway disease, vs β-blockers with CAD). Caution should be used when β-blockers are used with ND-CCBs. In patients with significant LV systolic dysfunction (LVEF ≤ 40%), maximally tolerated doses of evidence-based β-blockers (extended-release metoprolol succinate, bisoprolol, carvedilol) remain first-line therapy for rate control, although the benefits of adrenergic blockade, in addition to that provided by the control of the ventricular response rate, are uncertain.[583][588]

Recommendation

84. We recommend β-blockers or ND-CCBs (diltiazem or verapamil) be first-line agents for rate control of AF in patients without significant LV dysfunction (LVEF > 40%) (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the extensive clinical experience and record of safety and efficacy of β-blockers and ND-CCBs (verapamil and diltiazem) for AF rate control.

Practical Tip

The choice of specific rate-controlling agents should be guided by the patient’s characteristics and the drug efficacy/side effect profile.

Practical Tip

ND-CCBs (verapamil and diltiazem) have favourable pharmacological properties for rate control and might be the preferred choice in patients without a compelling indication for β-blocker usage.

85. We recommend evidence-based β-blockers (bisoprolol, carvedilol, metoprolol) be first-line agents for rate control of AF in patients with significant LV dysfunction (LVEF ≤ 40%) (Strong Recommendation; Moderate-Quality Evidence).

86. We recommend against rate control as a treatment strategy in patients with AF and who manifest preexcitation (Strong Recommendation; Moderate-Quality Evidence).

Practical Tip

See section 11.8 for the management of patients with AF and pre-excitation.

87. We suggest combination therapy (eg, a β-blocker with a ND-CCB) in patients who do not achieve satisfactory symptom or heart rate control with monotherapy (Weak Recommendation; Low-Quality Evidence).

Practical Tip

Combination therapy should be used with caution in patients at risk of significant bradycardia/AV block (eg, patients with resting sinus bradycardia or with significant conduction disease). These patients might require pacemaker implantation to facilitate pharmacological rate control.

Monotherapy with digoxin is generally ineffective in younger patients because of its inability to control ventricular rate during exertion or stress. Moreover, digoxin has a narrow therapeutic window, with observational evidence suggesting potential harmful effects when used for ventricular rate control.[589][592] However, a recent meta-analysis of 28 trials of digoxin for AF rate control showed no increase in all-cause mortality vs control intervention (RR, 0.82; 95% CI, 0.24-11.5).[593] As such, digoxin remains a reasonable choice for selected older or sedentary patients with HF and for those with inadequate rate control while receiving maximally tolerated doses of a β-blocker/ND-CCB. Although there is no direct evidence to support digoxin concentration monitoring for AF rate control, it might be reasonable to monitor patients at risk of digoxin-related adverse events (eg, female sex with low body weight and impaired renal function), at the clinician’s discretion, aiming for trough levels between 0.5 and 0.9 ng/mL.[594] Furthermore, it is important to note that coadministration of ND-CCBs and amiodarone will decrease digoxin clearance, resulting in a propensity toward toxicity.

Recommendation

88. We suggest that digoxin be considered as a monotherapy in older or sedentary individuals with permanent AF; or those with side effects or contraindications to first-line agents; or in addition to first-line agents in those who fail to achieve satisfactory symptom or heart rate control (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a lesser value on observational cohort studies in which adverse outcomes from digoxin have been reported.

Practical Tip

Digoxin is relatively ineffective for heart rate control in younger patients during activity but might be useful in older or sedentary individuals with HF, particularly in combination therapy.

Practical Tip

Therapeutic drug monitoring might be useful in adjusting digoxin dose, particularly in patients at risk of digoxin-related adverse events (eg, female sex with low body weight and impaired renal function). In patients with HF with reduced EF (HFrEF) trough levels between 0.5 and 0.9 ng/mL were associated with a significant decrease in all cause mortality and hospitalizations compared with levels ≥ 1.0 ng/mL, however, the optimal trough level for AF patients is unknown.

Amiodarone is a class III antiarrhythmic with complex pharmacological properties and potential serious adverse effects. However, selected patients such as the critically ill or those with side effects from, or contraindication to, first-line agents might benefit from amiodarone for rate control after careful consideration of alternative agents, alternative approaches (eg, transition to rhythm control), and risk/benefits of continued amiodarone therapy.[595],[596]

Recommendation

89. We recommend that amiodarone be used for AF rate control only in highly-selected patients such as the critically ill or those with significant side effects from or contraindication to first-line agents after careful consideration of alternative agents and risk/benefits of amiodarone therapy (Strong Recommendation; Low-Quality Evidence).

Dronedarone should not be used for AF rate control because it was associated with excess HF, stroke, and cardiovascular death in the Permanent Atrial Fibrillation Outcome Study Using Dronedarone on Top of Standard Therapy (PALLAS) trial.[597]

Recommendation

90. We recommend dronedarone not be used for AF rate control or in patients with HF (Strong Recommendation; High-Quality Evidence).

Values and Preferences

This recommendation places a high value on randomized controlled trial data that have shown that the use of dronedarone resulted in excess of cardiovascular death, and unplanned cardiovascular hospitalization.

9.2.2 Targets

The goals of ventricular rate control are the reduction of AF-related symptoms and the prevention of adverse cardiovascular events, rather than the achievement of a specific heart rate target. It is known that overly aggressive rate control is associated with adverse outcomes (eg, risk of symptomatic bradycardia with subsequent pacemaker implantation) and increased frequency of medical encounters. Conversely, overly lenient rate control might lead to HF (eg, tachycardia-mediated cardiomyopathy). In addition, specific populations might need stricter HR targets (eg, patients with cardiac resynchronization therapy [CRT], HF, tachycardia-mediated cardiomyopathy, mitral stenosis, stable angina), whereas others might do well with a more lenient target. As such, the intensity of AF rate control beyond the target HR of ≤ 100 bpm should be individualized on the basis of clinical characteristics and coexisting cardiovascular diagnoses. Most of the evidence to guide clinical decision-making for ventricular rate control targets has been acquired in patients with preserved LVEF.[512][514],[598] Previous guidelines have recommended heart rate targets of < 80 bpm at rest and < 110 bpm with exercise, because these targets were used in the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study.[512] However, retrospective analyses of the Rate Control versus Electrical Cardioversion for Persistent Atrial Fibrillation (RACE) and AFFIRM studies suggested that cardiovascular morbidity, mortality, and QOL did not differ between those achieving or not achieving the prespecified heart rate target (but still maintaining a resting heart rate < 100 bpm).[599] The prospective randomized Ratecontrol Efficacy in Permanent Atrial Fibrillation: a Comparison between Lenient Versus Strict Rate-control-II (RACE-II) trial598 showed that lenient rate control (resting heart rate target < 110 bpm) was noninferior to strict rate control (resting heart rate target < 80 bpm, < 110 bpm with exercise), with fewer medications, lower medication doses, fewer adverse events, and reduced health care utilization. However, it is important to recognize that the mean heart rates achieved were 76 ± 14 bpm in the strict group and 85 ± 14 bpm in the lenient group, with very few of those patients randomized to lenient rate control having resting heart rate of > 100 bpm.[600] As such, there remain some questions as to the ideal heart rate target. A recent analysis of the Outcomes Registry for Better Informed Treatment of Atrial Fibrillation (ORBITAF) registry and a retrospective analysis of the AFFIRM and Atrial Fibrillation and Congestive Heart Failure (AF-CHF) studies provides further insight. In both studies a U-shaped relationship between the resting heart rate and adverse outcomes was observed, with increased adverse event rates at the 2 extremes of resting heart rates. Although the combined AFFIRM/AF-CHF analysis suggested lower rates of mortality for those with resting heart rates in AF between 76 and 89 bpm, and lower rates of hospitalization for those with resting heart rates in AF between 76 and 114 bpm, the optimal heart rate in the ORBIT-AF registry was noted to be approximately 65 bpm.[601],[602] On the basis of the best available evidence, a heart rate-target of < 100 bpm at rest appears to be associated with an acceptable risk/benefit profile in patients without significant LV systolic dysfunction (LVEF > 40%). Because of the paucity of data to guide heart rate targets in patients with significant LV systolic dysfunction it is reasonable to target a heart rate of < 100 bpm, although clinically driven targets should be individualized to symptoms and hemodynamics as per in other populations.

Recommendation

91. We recommend titrating rate-controlling agents to achieve a resting heart rate of < 100 bpm during AF (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the small number of RCTs that have shown outcomes comparable between strict vs lenient strategies for AF rate control.

Practical Tip

The goal of AF rate control should be the control of AF-related symptoms and cardiovascular complications. Specific populations (eg, those with HF, tachycardiamediated cardiomyopathy, mitral stenosis, stable angina, patients with CRT) might need stricter HR targets whereas others might do well with a more lenient target.

Practical Tip

Paroxysmal AF and AFL can be more challenging to rate-control than persistent/permanent AF. Rhythm control should be considered for these patients.

92. We recommend maximizing evidence-based β-blocker dose in patients with reduced LVEF (LVEF ≤40%), in addition to achieving a resting heart rate of ≤ 100 bpm (Strong Recommendation; Moderate-Quality Evidence).

In patients with AF and a CRT device, the goals of care should be to maximize biventricular pacing (ie, as close to 100% as possible) rather than target a specific heart rate. If use of combination pharmacotherapy does not achieve a high percentage of biventricular pacing, then AV junction (AVJ) ablation should be considered.

Recommendation

93. We recommend that pharmacological atrioventricular blockade in patients with AF and CRT should target maximal biventricular pacing (as close to 100% as possible) and not a specific heart rate target (Strong Recommendation; High-Quality Evidence).

In patients with paroxysmal AF a rhythm control approach should be preferentially considered because ventricular rate control can be challenging. Finally, there is insufficient evidence to support routine monitoring of exercise heart rates. In the subset of very active individuals or patients with exercise-related symptoms it might be reasonable to monitor heart rate during exercise, and additionally target a heart rate of &lt; 110 bpm on moderate exertion (6-minute walk test) and a maximal heart rate of &lt; 110% of age-predicted maximum heart rate at peak exertion.

Recommendation

94. We suggest monitoring of the heart rate during exercise only in patients with exercise-related symptoms or in highly active individuals (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation places a high value on the lack of evidence to support heart rate monitoring during exercise.

9.2.3 Atrioventicular junction ablation and pacing

Pharmacological agents achieve satisfactory heart rate control in most patients; however, a subset of patients fail to achieve adequate control of ventricular rate despite maximally tolerated/combination doses of rate-controlling agents. Implantation of a permanent pacemaker followed by AVJ ablation can be a useful treatment strategy to achieve definitive rate control in this population with permanent AF. An AVJ ablation strategy ensures reliable control of the ventricular rate, regularization of the RR intervals, and discontinuation of rate/rhythm control drugs in most patients.[603] In patients with HF and CRT devices, AVJ ablation optimizes the delivery of biventricular pacing (see section 9.2.3.1). Compared with medical therapy, AVJ ablation and pacemaker implantation results in significant improvements in symptoms and QOL despite no significant changes in exercise capacity or functional status (eg, treadmill exercise or VO2 max).[604],[605] In general the greatest improvement in QOL is observed in patients who are able to discontinue rate-limiting pharmacotherapies.[606] It is important to note that these studies were performed in patients with permanent AF refractory to pharmacological rate control. As such, there are no data to support AVJ ablation and pacemaker implantation as a first-line treatment (ie, prior to attempts at pharmacological rate control). Patients with paroxysmal/persistent AF should be considered for a rhythm control strategy including catheter ablation (section 9.4) prior to pursuing AVJ ablation given the fact that AVJ ablation is irreversible, rendering patients pacemaker dependent, further complicating the management of device infection and lead failure, and increasing the risk of pacing induced cardiomyopathy.[603] In patients with permanent AF who undergo AVJ ablation, a single-chamber right ventricular pacemaker is preferred for those with normal ventricular function. In those with normal ventricular function CRT implantation before AVJ ablation has not been shown to result in substantial benefit over the single-chamber right ventricular pacemaker. In a meta-analysis of 4 trials comparing denovo CRT vs right ventricular pacemaker in patients with AF treated with AVJ ablation, CRT therapy resulted in only a small improvement in QOL (Minnesota Living with Heart Failure Questionnaire, 2.72 fewer points; 95% CI, 1.45-3.99) and LVEF (þ2.6%; 95% CI 1.69%-3.44%), at the expense of a trend toward increased procedure-related complications (RR, 1.96; 95% CI, 0.71-5.45; P ¼ 0.2).[607] Although there are emerging data on leadless pacemaker implantation and conduction system pacing (His bundle/left bundle branch area pacing) in the context of AVJ ablation, further studies are required before these strategies can be routinely recommended in preference to conventional single chamber right ventricular pacemakers.[608][610] For patients with LV systolic dysfunction being considered for AVJ ablation for permanent AF, a CRT device is preferable to a single-chamber right ventricular pacemaker. In susceptible patients chronic right ventricular pacing might lead to progressive LV dysfunction and HF. Biventricular pacing has been shown to significantly lower incidence of death or HF hospitalization (HR, 0.74; 95% CI, 0.60-0.90) compared with right ventricular pacing in patients with LVEF ≤ 50%.[611]

Recommendation

95. We recommend permanent pacemaker implantation with AVJ ablation in patients ineligible for rhythm control who have an uncontrolled heart rate during AF despite maximally tolerated combination pharmacological rate control (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the efficacy and safety of permanent pacemaker implantation with AVJ ablation for patients with refractory permanent AF.

Practical Tip

Patients with paroxysmal or persistent AF should be considered for a rhythm control strategy (eg, catheter ablation of AF) before proceeding with permanent pacemaker implantation with AVJ ablation.

96. We recommend against permanent pacemaker implantation with AVJ ablation in patients with an uncontrolled heart rate during AF without previous attempts at pharmacological rate control (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the lack of evidence to support a strategy of early pacemaker implantation and AVJ ablation (eg, before an adequate trial of pharmacological rate control), because of the longer-term consequences of iatrogenic pacemaker dependence.

9.2.3.1 HF, AF, and biventricular devices

HF and AF often coexist, with up to 25% of patients with a CRT device having coexisting AF.612 In this population, the outcomes of CRT are suboptimal with a larger proportion of “non-responders” and a higher mortality rate vs patients in sinus rhythm.[603] Therapeutic failure is largely due to the irregularity of the AF interfering with the ability of the device to deliver optimal CRT.[613],[614] Management options to optimize CRT include increasing the lower rate limit, uptitrating atrioventricular nodal-blocking drugs, and/or AVJ ablation. A meta-analysis of 6 studies that included 768 patients with AF, symptomatic HF, LVEF  35%, and a CRT device reported that AVJ ablation was associated with lower all-cause mortality (RR, 0.42; 95% CI, 0.26-0.68; P < 0.001), lower cardiovascular mortality (RR, 0.44; 95% CI, 0.24-0.81; P ¼ 0.008), greater improvement in NYHA class (0.34; 95% CI, 0.56 to 0.13; P ¼ 0.002), and a reduction in appropriate/ inappropriate defibrillator shocks, compared with medical therapy for ventricular response rate control.[615],[616] These potential benefits of AVJ ablation have to be weighed against the implications of rendering a patient pacemaker dependent. The Cardiac Resynchronisation Therapy and AV Nodal Ablation Trial in Atrial Fibrillation Patients (CAAN-AF; NCT01522898) and the Resynchronization/Defibrillation for Ambulatory Heart Failure Trial in Patients With Permanent AF (RAFT-Perm AF; NCT01994252) trials should provide additional insight into the role of CRT in patients with AF and HF.

Recommendation

97. We suggest AVJ ablation in HF patients with AF who are CRT nonresponders with biventricular pacing < 95% despite maximally tolerated doses of ratecontrolling drugs (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the documented importance of a high percentage of biventricular pacing for effective resynchronization therapy.

9.3 Long-term pharmacologic rhythm control

9.3.1 Antiarrhythmic drugs

A strategy of sinus rhythm maintenance using long-term antiarrhythmic drug therapy is preferred for those with recently diagnosed AF (ie, within a year), and might be considered for other symptomatic patients with established AF (Fig. 16). Because long-term antiarrhythmic therapy might not completely suppress AF, the focus of rhythm control should be on symptom relief, improving functional capacity and QOL, and reducing health care utilization while balancing potential adverse drug effects. Moreover, a recent study showed that an initial rhythm control strategy for patients with recently diagnosed AF was associated with decreased cardiovascular mortality and a reduced incidence of thromboembolic events compared with rate control alone.[515] Efficacy and safety data of common antiarrhythmic drugs used for rhythm control have been summarized in several systematic reviews.[617][619] In a meta-analysis of 59 RCTs, the pooled recurrence rates of AF was 64%-84% at 1 year in control participants. Antiarrhythmic therapy reduced recurrence rates to 20%-50%. The most effective drug was amiodarone (OR, 0.22; 95% CI, 0.16-0.29 for recurrence vs placebo).[617] Proarrhythmic events (ventricular or bradyarrhythmia) were significantly more frequent with sotalol (OR, 6.44; 95% CI, 1.03-40.24; P ¼ 0.047) and propafenone (OR, 4.06; 95% CI, 1.13-14.52; P ¼ 0.035), but were not significantly more frequent for flecainide (OR, 6.77; 95% CI, 0.85-54.02; P ¼ 0.067) or amiodarone (OR, 5.45; 95% CI, 0.69-42.93; P ¼ 0.095). Antiarrhythmic drugs have not been associated with a beneficial effect on mortality, and long-term use of sotalol and amiodarone have been associated with increased mortality (OR, 4.32; 95% CI, 1.59-11.70; P ¼ 0.013 and OR, 2.73; 95% CI, 1.00-7.41; P ¼ 0.049, respectively).[617] A single study showed stroke risk reduction with dronedarone use compared with placebo (OR, 0.69;95% CI, 0.57-0.84) however, this finding has not been confirmed by other studies.[620],[621] The antiarrhythmic drug doses, contraindications, or precautions are summarized in Supplemental Table S11. The initial choice of antiarrhythmic therapy is primarily driven by safety and tolerability, because these agents have a relatively similar efficacy (Fig. 19). If the initial drug does not achieve the desired results, an alternative antiarrhythmic may be used or catheter ablation may be considered. When the decision is made to abandon pharmacologic rhythm control and favour a rate control strategy, the antiarrhythmic drug should be discontinued.

Recommendation

98. We recommend a rhythm control strategy for patients with established AF who remain symptomatic with rate control therapy, or in whom rate control therapy is unlikely to control symptoms (Strong Recommendation; Moderate-Quality Evidence). We suggest consideration be given to rhythm control rather than rate control for patients with newly diagnosed AF (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation places a high value on the significant reductions in cardiovascular mortality and stroke observed in patients with newly diagnosed AF (within a year) treated with rhythm control, and lesser value on the increased adverse events observed in such patients. For those with established AF this recommendation places a high value on a decision-making process shared with patients, which considers the likelihood of improved symptoms, QOL, and health care utilization while minimizing adverse drug effects compared with other treatment strategies (rate control or catheter ablation).

Practical Tip

In select cases ablation might be preferred as first-line therapy (eg, rather than oral antiarrhythmic therapy) for patients with recurrent AF in whom long-term rhythm control is desired.

Practical Tip

Long-term oral antiarrhythmic therapy should not be continued in patients when AF becomes permanent.

99. We recommend that the goal of rhythm control therapy should be an improvement in cardiovascular outcomes, patient symptoms, and health care utilization, and not necessarily the elimination of all AF episodes (Strong Recommendation; Moderate-Quality Evidence).

100. We recommend that the choice of antiarrhythmic drug used for long-term pharmacologic rhythm control be defined according to patient characteristics (Strong Recommendation; Moderate-Quality Evidence).

101. We recommend that pharmacologic rhythm control should be avoided in patients with AF and advanced sinus or AV nodal disease unless the patient has a pacemaker or implantable defibrillator (Strong Recommendation; Low-Quality Evidence).

102. We recommend an AV nodal-blocking agent (β-blockers and ND-CCBs) be used in combination with class I antiarrhythmic drugs (eg, flecainide or propafenone) (Strong Recommendation; Low-Quality Evidence).

103. We recommend the use of amiodarone for pharmacologic rhythm control only when the potential for drug toxicities is considered and when other choices are contraindicated or have failed (Strong Recommendation; Low-Quality Evidence).

9.3.1.1 Flecainide and propafenone

Class Ic antiarrhythmic agents, such as flecainide and propafenone, are use-dependent sodium channel-blocking drugs.[622] The concomitant use of AV nodal blocking agents is recommended with class Ic antiarrhythmic drug therapy because of the potential risk of AF organization into AFL, with the potential of 1:1 AV conduction and rapid ventricular rate. Class Ic agents should be avoided in patients with: (1) preexisting advanced AV block (second- or third-degree AV block) or significant conduction system disorders (left bundle branch block, or right bundle branch block when associated with left hemiblock); (2) LV systolic dysfunction (LVEF ≤ 40%); (3) significant LV hypertrophy; (4) severe hepatic or severe renal impairment (CrCl < 35 mL/min); and (5) ischemic heart disease (active ischemia or history of MI). Because the use of flecainide has been associated with increased mortality when administered to suppress ventricular ectopy in the context of recent MI,[623] a formal ischemia assessment (eg, stress test) should be considered before initiation of class Ic antiarrhythmic drugs in patients older than 50 years of age or with significant atherosclerotic risk factors. In addition, it is reasonable to consider annual assessment of symptoms of CAD for patients receiving long-term class Ic antiarrhythmic use, with formal stress testing being performed if significant symptoms are present. An ECG should be performed at baseline and after initiation to monitor for PR and QRS interval prolongation. An increase in QRS duration > 25% compared with baseline increases proarrhythmia risk.[624]

9.3.1.2 Sotalol

Sotalol is a drug with reverse use dependence Ikr inhibition and is also a β-blocker. At lower doses the β-blocker effects predominate whereas the class III effects emerge with higher doses. The major risk of sotalol is QT prolongation and TdP. Sotalol should be avoided in patients with: (1) preexisting QTc prolongation (congenital or acquired long QT syndromes); (2) high-degree AV conduction disorders; (3) severe renal impairment (dose adjustment required for CrCl 40-60 mL/min; avoid with CrCl < 40 mL/min); (4) significant LV systolic dysfunction (LVEF ≤ 40%); or (5) significant risk factors for TdP (eg, women aged older than 65 years who are receiving diuretics or those with renal insufficiency). An ECG should be performed at baseline and 48-72 hours after outpatient initiation to monitor for QT interval prolongation.

9.3.1.3 Amiodarone

Amiodarone is a multichannel blocker and a nonselective β-blocker. A loading regimen of 10-12 g is recommended and then maintenance of ≤ 200 mg daily. Amiodarone has a long half-life and long-term use increases the risk for numerous extracardiac toxicities that affect the skin, thyroid, pulmonary, liver, and neurological systems. It should not be used as a firstline therapy when another drug might be an option. Amiodarone should be avoided in patients with: (1) high-degree AV conduction disorders; (2) active hepatitis or significant chronic liver disease; (3) pulmonary interstitial abnormalities; (4) preexisting QTc prolongation (congenital or acquired long QT syndromes); (5) hypersensitivity to the drug components, including iodine; or (6) concomitant use of strong cytochrome P450 3A4 (CYP3A4) inhibitors (eg, ketoconazole, cyclosporin, clarithromycin, ritonavir). Surveillance investigations are recommended with the use of amiodarone (eg, liver and thyroid tests every 6 months, annual chest radiograph).[625] Amiodarone inhibits CYP3A, CYP2C9, and P-glycoprotein drugs, which requires close monitoring and dose adjustment of other affected medications (eg, digoxin, VKA, HMG-CoA reductase inhibitors). Patients should be counselled to be diligent in use of sun protection because of photosensitivity and report any symptoms indicative of pulmonary toxicity (eg, persistent nonproductive cough), optic neuropathy (changes in visual acuity and decreases in peripheral vision), or hepatic injury.

9.3.1.4 Dronedarone

Dronedarone resembles amiodarone but removal of iodine and the addition of a methane-sulfonyl group results in shortening the half-life (approximately 24 hours) and less tissue accumulation.[626] Dronedarone is the only antiarrhythmic drug shown to reduce hospitalization and cardiovascular mortality in patients with paroxysmal or persistent AF with at least 1 additional risk factor for death.[620] Dronedarone should be avoided in patients with: (1) permanent AF; (2) HF with recent decompensation requiring hospitalization or LV systolic dysfunction (LVEF ≤ 40%), owing to the observed increase in mortality observed with dronedarone use in this population597; (3) high-degree AV conduction disorders; (4) patients with previous lung or liver injury related to previous use of amiodarone; (5) preexisting QTc prolongation; or (6) severe hepatic impairment. Because of the risk of hepatotoxicity it is recommended that liver function tests be performed every 3 months for the first year of use, then every 6 months thereafter.

9.3.2 Pill-in-the-pocket antiarrhythmic drug therapy

In selected patients with symptomatic, infrequent, longer-lasting episodes of AF, the use of intermittent class Ic antiarrhythmic therapy (“pill-in-the-pocket”) shortly after symptom onset to restore sinus rhythm might be an alternative approach to daily antiarrhythmic use.[557],[560][563] Indications, contraindications, and monitoring details are presented in Supplemental Table S12.

Recommendation

104. We recommend intermittent antiarrhythmic drug therapy (“pill-in-the-pocket”) as an alternative to daily antiarrhythmic therapy in patients with infrequent, symptomatic episodes of AF (Strong Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation is on the basis of the results of observational cohort studies that have shown efficacy and safety of intermittent antiarrhythmic drug therapy in selected patients. It places a high value on patient preferences and capabilities.

9.3.3 Trial of cardioversion

Many patients with persistent AF can present with the insidious onset of vague symptoms, such as fatigue or decreased exercise tolerance. In this population it can be difficult to determine the contribution of AF to their clinical presentation. A trial of sinus rhythm using cardioversion can often be useful in this population with established AF to determine if these nonspecific symptoms are secondary to AF. If so, then rhythm control might be the preferred treatment strategy (Fig. 16).

9.4 Catheter ablation of AF

Catheter ablation of AF has emerged as an important therapeutic modality for this common arrhythmia. The goal of catheter ablation is to eliminate the triggers and substrate responsible for the initiation and maintenance of AF. Ablation is performed as a percutaneous procedure in which catheters are inserted through venous access into the heart using fluoroscopic and electroanatomical mapping systems to identify regions of interest. These regions can then be ablated using thermal energy, namely radiofrequency or cryothermy.[627] Irreversible electroporation (pulsed electrical fields) is a recent modality that might achieve the same effect, without the risk of thermal energy-related complications.[628] Most of the triggers for AF originate within the PVs of the left atrium.[629] Therefore, the “cornerstone” of all AF ablation procedures is ablating around the PV to electrically isolate them (Fig. 20). PV isolation (PVI) for paroxysmal AF is associated with success rates ranging from 60% to 80% after 1 procedure. For persistent AF, single-procedure success rates are lower (50%-70%), and it is unclear if ablation beyond PVI is required.[630] Although the success rates seem suboptimal, these are often determined on the basis of the total elimination of AF. However, in many cases substantial clinical benefit can be achieved without total elimination of AF.[631] When considered quantitatively, AF ablation substantially reduces the burden of AF (ie, time spent in arrhythmia) by > 98%.[627] The CABANA trial (2204 patients) is the largest trial of catheter ablation vs medical therapy.[394] In this trial patients with previous antiarrhythmic drug failure (47%) as well as antiarrhythmic-naive patients (53%) were enrolled. Although in the intention to treat analysis the trial did not show a difference between ablation and medical therapy for the primary end point of death, stroke, bleeding, and cardiac arrest, there was a significant reduction in the secondary end point of mortality and cardiovascular hospitalization in the ablation group (HR, 0.83; 95% CI, 0.74-0.93; P ¼ 0.001). Moreover, ablation was associated with a significant reduction in AF recurrence (HR, 0.52; 95% CI, 0.45-0.60; P < 0.001) with the per-protocol analysis showing that ablation was associated with a significant reduction in the primary end point (HR, 0.73; 95% CI, 0.54-0.99; P ¼ 0.046).

9.4.1 Procedural considerations for catheter ablation of AF

AF ablation is a complex procedure that requires a high degree of operator expertise. Recent balloon-based technologies have helped to make the procedures shorter and more consistent, but there remains evidence that such procedures should only be performed by individuals with appropriate experience and technological support. Recent evidence has shown that the rate of complications is directly related to the number of procedures performed at an institution and/or by the operator.[632],[633]

Recommendation

105. We suggest that catheter ablation of AF should be performed by electrophysiologists with a high degree of expertise and high annual procedural volumes (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation recognizes that the risks of catheter ablation are directly related to operator experience and procedural volume at a given centre. Although it is difficult to specify exact numerical values, the threshold seems to be 25-50 procedures per operator per year.

9.4.2 Catheter ablation of AF after a trial of antiarrhythmic drugs

The primary indication for AF ablation is to achieve rhythm control in patients in whom antiarrhythmic drug therapy has already failed. Two systematic reviews have been performed, one before (2272 patients in 17 RCTs)[634] and one including CABANA (4464 patients in 18 RCTs).[635] Overall, the quality of studies was high and risk of bias low. Most patients included in these trials were patients in whom at least 1 antiarrhythmic drug had already failed. Despite the large difference in sample size, outcomes were remarkably consistent with reasonably large reductions in cardiovascular hospitalizations (RR, 0.63; 95%CI, 0.46-0.87; P ¼ 0.01, and RR, 0.56; 95% CI, 0.39-0.81; P ¼ 0.0001, respectively) and AF recurrences (RR, 0.44; 95% CI, 0.31-0.61; P ¼ 0.001, and RR, 0.42; 95% CI, 0.33-0.53; P < 0.00001, respectively) among patients who underwent catheter ablation, compared with antiarrhythmic therapy. All-cause mortality was also reduced in both meta-analyses but was driven by studies that enrolled patients with HF and reduced EF; sensitivity analysis showed no statistically significant reduction in all-cause mortality when these studies were removed from the analysis (RR, 0.48; 95% CI, 0.23-1.01; P ¼ 0.05, and RR, 0.67; 95% CI, 0.23-1.99; P ¼ 0.47, respectively). In patients without HF, mortality rates were too low to assess the potential for mortality benefit. Stroke rates were very low across all study populations and no effect of catheter ablation was shown on the rate of stroke (0.68% vs 1.23% for medical therapy; RR, 0.56; 95% CI, 0.26- 1.22; P ¼ 0.14). Likewise, major bleeding was not significantly more frequent after catheter ablation (3.4% vs 2.5% for medical therapy; RR, 1.55; 95% CI, 0.83-2.91; P ¼ 0.17). One RCT specifically addressed QOL as the primary outcome in patients in whom previous antiarrhythmic drug therapy had failed.636 Similar to contemporary trials,[631] it showed important improvements in AF-specific and general QOL driven predominantly by improvements in AF symptoms and AF-related hospitalizations.

Recommendation

106. We recommend catheter ablation of AF in patients who remain symptomatic after an adequate trial of antiarrhythmic therapy and in whom a rhythm control strategy remains desired (Strong Recommendation; High-Quality Evidence).

Values and Preferences

This recommendation recognizes the positive effect of catheter ablation on AF burden, symptoms, QOL, and cardiovascular hospitalizations, as well as the declining risks of the procedure.

Practical Tip

Catheter ablation might be the preferred means to maintain sinus rhythm in select patients with symptomatic AF and mild-moderate structural heart disease, particularly systolic HF, who are refractory or intolerant to 1 antiarrhythmic medication.

9.4.3 Catheter ablation of AF as first-line treatment

Early intervention for AF can prevent progression to persistent AF and avoid some of the long-term risks of the arrhythmia including stroke and HF. Furthermore, studies have shown that success rates for catheter ablation are higher when used earlier in the disease.[637] Therefore, several clinical trials have addressed whether AF ablation should be used as first-line therapy before the use of antiarrhythmic drugs. Three previous randomized studies suggested a benefit of first-line ablation over antiarrhythmic drugs but these were small.[638][640] More recently the large randomized Early Agressive Invasive Intervention for Atrial Fibrillation (EARLY-AF) trial demonstrated that first-line cryoballoon catheter ablation resulted in a signficant reduction in arrhythmia recurrence and AF burden, and a significant improvement in quality of life relative to first line antiarrhythmic drugs.[641]

Recommendation

107. We suggest catheter ablation to maintain sinus rhythm as first-line therapy for relief of symptoms in select patients with symptomatic AF (Weak Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation recognizes that patients might have relative or absolute contraindications to pharmacologic rhythm control.

9.4.4 Candidates for AF ablation

Similar to long-term antiarrhythmic drug therapy, the decision to pursue a strategy of sinus rhythm maintenance should be aimed primarily at reduction of patient symptoms to improve QOL and reduce health care utilization. Patients who have truly asymptomatic AF are not generally considered candidates for ablation, however ablation might be pursued in those in whom AF is thought to adversely affect LV function even in the absence of overt symptoms. A specific subgroup that deserves mention are patients with HF, in whom AF is a causative or major contributing factor. A number of studies and systematic reviews have shown clinically important improvements in HRQOL, exercise tolerance, and LV function associated with catheter ablation over pharmacological rate control or rhythm control strategies.[642],[643] In addition, 2 RCTs, including the Catheter Ablation vs Standard Conventional Therapy in Patients With Left Ventricular Dysfunction and Atrial Fibrillation (CASTLE-AF) trial, have recorded reduced hospitalizations in patients with HFrEF who underwent catheter ablation compared with medical therapy.[644],[645] A recent systematic review showed that catheter ablation in this population was associated with a statistically significant reduction in all-cause mortality (5.3% vs 7.9% for medical therapy; RR, 0.69; 95% CI, 0.54-0.88; P ¼ 0.003; 9 studies, 3576 patients).[635] Although a low level of bias was seen, the mortality benefit was driven by a single study (CASTLE-AF) that exclusively enrolled patients with HFrEF.[645] A recent observational study showed that catheter ablation was associated with a long-term reduction in all-cause mortality and HF rehospitalizations.[646] There are no data showing a mortality benefit in patients with HF and preserved EF. The large multicentre Canadian randomized ablation-based Randomized Ablation-Based Atrial Fibrillation Rhythm Control vs Rate Control Trial in Patients With Heart Failure and High Burden Atrial Fibrillation (RAFT-AF) study has closed enrollment and is due to report later this year (NCT01420393). These data will provide considerable insight into the effect of catheter ablation on HF with preserved EF and HFrEF. First-line therapy might be considered in younger patients who wish to avoid the risks of long-term antiarrhythmic agent use. Patients might also have cardiac or noncardiac absolute or relative contraindications to antiarrhythmic drugs. Some patients with tachy-brady syndrome are unable to tolerate drug therapy because of bradycardia complications in the absence of a pacemaker. If the AF can be successfully ablated, then antiarrhythmic therapy and the need for permanent pacing might be avoided.[647]

9.4.5 Catheter ablation of AFL

Although AF and AFL might coexist in the same patient, AFL is an arrhythmia that is distinct from AF. In contrast to the apparent disorganization of AF, AFL usually involves a single macroreentrant atrial circuit rotating around a large central functional or anatomic obstacle (valves, veins, scar). AFL can be classified as “typical” (cavo-tricuspid isthmus-dependent) or “atypical” (eg, non-cavo tricuspid isthmus-dependent right or left AFL). Atypical AFL arising from scar related to previous heart surgery (eg, atriotomy or prosthesis) or catheter ablation and is also known as “incisional” flutter. Catheter ablation of typical right AFL is preferred to pharmacological therapy because of the relatively high success rate and relatively low rate of periprocedural complications. Ablation is typically performed with the patient under conscious sedation, using the femoral approach, guided by fluoroscopy or 3-D electroanatomic mapping. The goal is to create a complete ablation line along the cavo-tricuspid isthmus between the tricuspid annulus and the inferior vena cava to achieve bidirectional electrical conduction block. This intervention has an excellent acute and long-term success rate 90%, which is more effective than pharmacological rhythm control.[400],[648],[649] In addition, ablation improves QOL, reduces symptoms, and might also avoid the development of a tachycardia-mediated cardiomyopathy.[648] The acute complication rate is 2.6%, most commonly related to vascular access problems.[400] Major complications, like complete heart block or pericardial effusion, are uncommon.[649] In patients with coexisting AF and AFL it is perfectly cromulent to consider a hybrid treatment approach, in which AFL ablation is performed to prevent recurrent AFL while antiarrhythmic drug therapy is used to control AF.[650] This approach has been effective for patients who derive clinical benefit from class Ic antiarrhythmic treatment of AF but manifest recurrent AFL.

Recommendation

108. We recommend catheter ablation of typical right AFL as a reasonable alternative to pharmacologic rhythm or rate control therapy (Strong Recommendation; Moderate-Quality Evidence).

Values and Preferences

This recommendation recognizes the high success rate and low complication rate of typical right AFL ablation, as well as the observation that typical right AFL is challenging to control medically with antiarrhythmic drugs and adequate rate control is difficult to achieve.

9.5 Arrhthmia surgery

Surgical treatment for AF can be accomplished as a “standalone” procedure or performed coincident with a planned surgical procedure (valve replacement/repair and/or CABG). A number of factors need to be considered when contemplating surgical AF ablation therapy, including the indication for the procedure (eg, the potential benefits of sinus rhythm), the likely efficacy of the procedure (considering local surgeon/institutional experience), and the safety of the procedure. Moreover, in the context of adjuvant surgical ablation (eg, surgical ablation at the time of another surgery) the type of concomitant cardiac surgery (eg, mitral valve, which necessitates left atriotomy, vs CABG surgery, which does not) needs to be considered. Recent studies have shown that LA ablation in addition to valve surgery and/or CABG was beneficial in terms of sinus rhythm maintenance.[651],[652] In the first study, 224 patients who underwent CABG and valve surgery were randomized to LA cryoablation or control; 60.2% of cryoablation patients showed sinus rhythm according to Holter monitoring at 1 year, vs 35.5% (P ¼ 0.002) of patients without ablation.[651] In the second study 63.2% of 260 mitral valve surgery patients randomized to ablation (either PVI alone or biatrial ablation [secondary randomization]) were in sinus rhythm at 1 year vs 29.4% of those randomized to no ablation (P < 0.001).[652] A recent systematic review combined 9 small RCTs and showed a large effect on the maintenance of sinus rhythm at 12 months in 481 patients (OR, 10.41; 95% CI, 5.30-20.44) as well as beyond 12 months (OR, 11.61; 95% CI, 4.53-29.79; 4 studies, 154 patients).[653] Although no heterogeneity was observed among these trials, no individual study randomized more than 95 patients. Despite the high reported rates of sinus rhythm maintenance after surgical ablation, the effect on other outcomes is controversial. Although there is evidence for an improvement in HRQOL outcomes with a surgical procedure that includes concomitant surgical AF ablation, in several RCTs the magnitude of improvement was observed to be similar to patients who did not undergo ablation at the time of cardiac surgery.[654][656] Moreover, surgical ablation appears to have no effect on the incidence of late stroke/TIA (6 RCTs; OR, 1.01; 95% CI, 0.41-2.49; P ¼ 0.98) or long-term survival (15 RCTs; OR, 0.91; 95% CI, 0.59-1.41; P ¼ 0.67).[657] A significant concern with surgical ablation relates to the possibility of increased postoperative morbidity secondary to the additional surgical procedure. In a large meta-analysis, concomitant surgical AF ablation was not associated with an increase in the incidence of perioperative morbidity (which included deep sternal wound infection, pneumonia, reoperation for bleeding, and renal failure) or perioperative ICU length of stay.[657] Likewise, there was no difference in the rate of readmission within 30 days, short-term survival (< 30 days), or perioperative stroke/TIA (< 30 days).[653],[657] However, as highlighted in the two aforementioned RCTs, surgical ablation was associated with a significant increase in the rates of pacemaker implantation (6% vs 1% in Budera et al.[651] and 21.5% vs 8.1% in Gillinov et al.[652]).

Recommendation

109. We suggest that a surgical AF ablation procedure be considered in association with a planned cardiac surgical procedure (eg, mitral valve, aortic valve, or coronary artery bypass surgery) in patients with symptomatic nonpermanent AF when the likelihood of success is deemed to be high, the additional risk is low, and sinus rhythm is expected to achieve substantial symptomatic benefit (Weak Recommendation; Low-Quality Evidence).

Values and Preferences

This recommendation recognizes that there is no evidence that surgical AF ablation influences hard outcomes (eg, stroke, mortality, thromboembolic complications). Patient considerations and individualized risk-benefit analyses should determine for whom the surgical procedure is performed.

Practical Tip

The symptomatic benefit of sinus rhythm needs to be balanced with the attendant risks of ablation surgery, including the increased need for permanent pacing (particularly for biatrial and/or Maze procedures).

There has been recent interest in hybrid AF ablation, achieved through collaboration between surgeons and electrophysiologists. This procedure encompasses surgical ablation (usually minimally invasive) coupled with percutaneous endocardial ablation of any residual gaps 6-8 weeks later. Similar to all invasive procedures, centre expertise and operator experience are important determinants of success. Comparative meta analyses suggest a modestly improved freedom from antiarrhythmic drug use after hybrid AF ablation in comparison to traditional Cox-Maze procedures, but former approach is associated with a higher rate of complications.[657],[658] Some have advocated for the hybrid approach to involve surgical bilateral PVI with LAA closure coupled with endocardial ablation protocols, although this approach has not been tested in an RCT. Often a bilateral thoracoscopic intervention is needed in this setting. Other hybrid approaches might include unilateral thoracoscopic PVI encircling box lesions and posterior LA wall epicardial ablation lesion sets without concomitant LAAO.[657] Although stand-alone surgical ablation presently comprises a small percentage of AF ablations, the ability to offer these newer approaches in a minimally invasive fashion without sternotomy or cardiopulmonary bypass is potentially attractive. Nevertheless, these approaches will need to be assessed in outcome studies.

Recommendation

110. We suggest that stand-alone surgical or hybrid ablation of AF may be considered for patients with symptomatic nonpermanent AF that is refractory to attempts at percutaneous catheter ablation and whose symptoms warrant the additional risk of a surgical procedure (Weak Recommendation; LowQuality Evidence).

References

505. Shaver CM, Chen W, Janz DR, et al. Atrial fibrillation is an independent predictor of mortality in critically ill patients. Crit Care Med 2015;43:2104-11.

506. Chen AY, Sokol SS, Kress JP, Lat I. New-onset atrial fibrillation is an independent predictor of mortality in medical intensive care unit patients. Ann Pharmacother 2015;49:523-7.

507. Yoshida T, Fujii T, Uchino S, Takinami M. Epidemiology, prevention, and treatment of new-onset atrial fibrillation in critically ill: a systematic review. J Intensive Care 2015;3:19.

508. Wetterslev M, Haase N, Hassager C, et al. New-onset atrial fibrillation in adult critically ill patients: a scoping review. Intensive Care Med 2019;45:928-38.

509. Chean CS, McAuley D, Gordon A, Welters ID. Current practice in the management of new-onset atrial fibrillation in critically ill patients: a UK-wide survey. PeerJ 2017;5:e3716.

510. Scheuermeyer FX, Pourvali R, Rowe BH, et al. Emergency department patients with atrial fibrillation or flutter and an acute underlying medical illness may not benefit from attempts to control rate or rhythm. Ann Emerg Med 2015;65:511-522.e512.

511. Stiell IG, Scheuermeyer FX, Vadeboncoeur A, et al. CAEP acute atrial fibrillation/flutter best practices checklist. CJEM 2018;20:334-42.

512. Wyse DG, Waldo AL, DiMarco JP, et al. A comparison of rate control and rhythm control in patients with atrial fibrillation. N Engl J Med 2002;347:1825-33.

513. Van Gelder IC, Hagens VE, Bosker HA, et al. A comparison of rate control and rhythm control in patients with recurrent persistent atrial fibrillation. N Engl J Med 2002;347:1834-40.

514. Roy D, Talajic M, Nattel S, et al. Rhythm control versus rate control for atrial fibrillation and heart failure. N Engl J Med 2008;358:2667-77.

515. Kirchhof P, Camm AJ, Goette A, et al. Early rhythm-control therapy in patients with atrial fibrillation. N Engl J Med 2020;383:1305-16.

516. Parkash R, Magee K, McMullen M, et al. The Canadian Community Utilization of Stroke Prevention Study in Atrial Fibrillation in the Emergency Department (C-CUSP ED). Ann Emerg Med 2019;73:382-92.

517. Atzema CL, Fang J, Cox JL, et al. Assessment of an algorithm for prescription of oral anticoagulation for patients with atrial fibrillation in emergency departments. JAMA Netw Open 2020;3:e200306.

518. Atzema CL, Yu B, Schull MJ, et al. The association of follow-up care with long-term death and subsequent hospitalization in patients with atrial fibrillation who receive emergency care in the province of Ontario. Circ Arrhythm Electrophysiol 2019;12:e006498.

519. Hawkins NM, Scheuermeyer FX, Youngson E, et al. Impact of cardiology follow-up care on treatment and outcomes of patients with new atrial fibrillation discharged from the emergency department. Europace 2020;22:695-703.

520. Atzema CL, Austin PC. Rate-control with beta-blockers versus calciumchannel blockers in the emergency setting: predictors of medication class choice and associated hospitalization. Acad Emerg Med 2017;24:1334-48.

521. Demircan C, Cikriklar HI, Engindeniz Z, et al. Comparison of the effectiveness of intravenous diltiazem and metoprolol in the management of rapid ventricular rate in atrial fibrillation. Emerg Med J 2005;22:411-4.

522. Fromm C, Suau SJ, Cohen V, et al. Diltiazem vs metoprolol in the management of atrial fibrillation or flutter with rapid ventricular rate in the emergency department. J Emerg Med 2015;49:175-82.

523. Martindale JL, deSouza IS, Silverberg M, Freedman J, Sinert R. bBlockers versus calcium channel blockers for acute rate control of atrial fibrillation with rapid ventricular response: a systematic review. Eur J Emerg Med 2015;22:150-4.

524. Hines MC, Reed BN, Ivaturi V, et al. Diltiazem versus metoprolol for rate control in atrial fibrillation with rapid ventricular response in the emergency department. Am J Health Syst Pharm 2016;73:2068-76.

525. Lip GY, Apostolakis S. Atrial fibrillation (acute onset). BMJ Clin Evid 2014;2014:0210.

526. Jandali MB. Safety of intravenous diltiazem in reduced ejection fraction heart failure with rapid atrial fibrillation. Clin Drug Investig 2018;38:503-8.

527. Hirschy R, Ackerbauer KA, Peksa GD, O’Donnell EP, DeMott JM. Metoprolol vs diltiazem in the acute management of atrial fibrillation in patients with heart failure with reduced ejection fraction. Am J Emerg Med 2019;37:80-4.

528. Goldenberg IF, Lewis WR, Dias VC, Heywood JT, Pedersen WR. Intravenous diltiazem for the treatment of patients with atrial fibrillation or flutter and moderate to severe congestive heart failure. Am J Cardiol 1994;74:884-9.

529. Stratton T, Nasser L. BET 1: lenient or strict rate control for atrial fibrillation. Emerg Med J 2018;35:765-8.

530. Dorian P, Angaran P. b-Blockers and atrial fibrillation: hypertension and other medical conditions influencing their use. Can J Cardiol 2014;30:S38-41.

531. Michael JA, Stiell IG, Agarwal S, Mandavia DP. Cardioversion of paroxysmal atrial fibrillation in the emergency department. Ann Emerg Med 1999;33:379-87.

532. Slavik RS, Tisdale JE, Borzak S. Pharmacologic conversion of atrial fibrillation: a systematic review of available evidence. Prog Cardiovasc Dis 2001;44:121-52.

533. Burton JH, Vinson DR, Drummond K, et al. Electrical cardioversion of emergency department patients with atrial fibrillation. Ann Emerg Med 2004;44:20-30.

534. Coll-Vinent B, Fuenzalida C, Garcia A, Martin A, Miro O. Management of acute atrial fibrillation in the emergency department: a systematic review of recent studies. Eur J Emerg Med 2013;20:151-9.

535. Stiell IG, Clement CM, Rowe BH, et al. Outcomes for emergency department patients with recent-onset atrial fibrillation and flutter treated in Canadian hospitals. Ann Emerg Med 2017;69:562-571.e562.

536. Madrid AH, Moro C, Marin-Huerta E, et al. Comparison of flecainide and procainamide in cardioversion of atrial fibrillation. Eur Heart J 1993;14:1127-31.

537. Kochiadakis GE, Igoumenidis NE, Hamilos ME, et al. A comparative study of the efficacy and safety of procainamide versus propafenone versus amiodarone for the conversion of recent-onset atrial fibrillation. Am J Cardiol 2007;99:1721-5.

538. Stiell IG, Sivilotti MLA, Taljaard M, et al. Electrical versus pharmacological cardioversion for emergency department patients with acute atrial fibrillation (RAFF2): a partial factorial randomised trial. Lancet 2020;395:339-49.

539. Priori SG, Wilde AA, Horie M, et al. HRS/EHRA/APHRS expert consensus statement on the diagnosis and management of patients with inherited primary arrhythmia syndromes: document endorsed by HRS, EHRA, and APHRS in May 2013 and by ACCF, AHA, PACES, and AEPC in June 2013. Heart Rhythm 2013;10:1932-63.

540. Stambler BS, Wood MA, Ellenbogen KA, et al. Efficacy and safety of repeated intravenous doses of ibutilide for rapid conversion of atrial flutter or fibrillation. Ibutilide Repeat Dose Study Investigators. Circulation 1996;94:1613-21.

541. Volgman AS, Carberry PA, Stambler B, et al. Conversion efficacy and safety of intravenous ibutilide compared with intravenous procainamide in patients with atrial flutter or fibrillation. J Am Coll Cardiol 1998;31:1414-9.

542. Abi-Mansour P, Carberry PA, McCowan RJ, et al. Conversion efficacy and safety of repeated doses of ibutilide in patients with atrial flutter and atrial fibrillation. Study Investigators. Am Heart J 1998;136:632-42.

543. Reisinger J, Gatterer E, Lang W, et al. Flecainide versus ibutilide for immediate cardioversion of atrial fibrillation of recent onset. Eur Heart J 2004;25:1318-24.

544. Simon A, Niederdoeckl J, Skyllouriotis E, et al. Vernakalant is superior to ibutilide for achieving sinus rhythm in patients with recent-onset atrial fibrillation: a randomized controlled trial at the emergency department. Europace 2017;19:233-40.

545. Vogiatzis I, Papavasiliou E, Dapcevitch I, Pittas S, Koulouris E. Vernakalant versus ibutilide for immediate conversion of recent-onset atrial fibrillation. Hippokratia 2017;21:67-73.

546. Vinson DR, Lugovskaya N, Warton EM, et al. Ibutilide effectiveness and safety in the cardioversion of atrial fibrillation and flutter in the community emergency department. Ann Emerg Med 2018;71:96-108.e102.

547. Zhang N, Guo JH, Zhang H, et al. Comparison of intravenous ibutilide vs propafenone for rapid termination of recent onset atrial fibrillation. Int J Clin Pract 2005;59:1395-400.

548. Kafkas NV, Patsilinakos SP, Mertzanos GA, et al. Conversion efficacy of intravenous ibutilide compared with intravenous amiodarone in patients with recent-onset atrial fibrillation and atrial flutter. Int J Cardiol 2007;118:321-5.

549. Tercius AJ, Kluger J, Coleman CI, White CM. Intravenous magnesium sulfate enhances the ability of intravenous ibutilide to successfully convert atrial fibrillation or flutter. Pacing Clin Electrophysiol 2007;30:1331-5.

550. Patsilinakos S, Christou A, Kafkas N, et al. Effect of high doses of magnesium on converting ibutilide to a safe and more effective agent. Am J Cardiol 2010;106:673-6.

551. Stiell IG, Dickinson G, Butterfield NN, et al. Vernakalant hydrochloride: a novel atrial-selective agent for the cardioversion of recent-onset atrial fibrillation in the emergency department. Acad Emerg Med 2010;17:1175-82.

552. Camm AJ, Toft E, Torp-Pedersen C, et al. Efficacy and safety of vernakalant in patients with atrial flutter: a randomized, double-blind, placebo-controlled trial. Europace 2012;14:804-9.

553. McIntyre WF, Healey JS, Bhatnagar AK, et al. Vernakalant for cardioversion of recent-onset atrial fibrillation: a systematic review and meta-analysis. Europace 2019;21:1159-66.

554. Stiell IG, Roos JS, Kavanagh KM, Dickinson G. A multicenter, openlabel study of vernakalant for the conversion of atrial fibrillation to sinus rhythm. Am Heart J 2010;159:1095-101.

555. Pratt CM, Roy D, Torp-Pedersen C, et al. Usefulness of vernakalant hydrochloride injection for rapid conversion of atrial fibrillation. Am J Cardiol 2010;106:1277-83.

556. Roy D, Pratt CM, Torp-Pedersen C, et al. Vernakalant hydrochloride for rapid conversion of atrial fibrillation: a phase 3, randomized, placebo-controlled trial. Circulation 2008;117:1518-25.

557. Khan IA, Mehta NJ, Gowda RM. Amiodarone for pharmacological cardioversion of recent-onset atrial fibrillation. Int J Cardiol 2003;89:239-48.

558. Alp NJ, Bell JA, Shahi M. Randomised double blind trial of oral versus intravenous flecainide for the cardioversion of acute atrial fibrillation. Heart 2000;84:37-40.

559. Boriani G, Capucci A, Lenzi T, Sanguinetti M, Magnani B. Propafenone for conversion of recent-onset atrial fibrillation. A controlled comparison between oral loading dose and intravenous administration. Chest 1995;108:355-8.

560. Khan IA. Single oral loading dose of propafenone for pharmacological cardioversion of recent-onset atrial fibrillation. J Am Coll Cardiol 2001;37:542-7.

561. Alboni P, Botto GL, Baldi N, et al. Outpatient treatment of recentonset atrial fibrillation with the “pill-in the-pocket” approach. N Engl J Med 2004;351:2384-91.

562. Saborido CM, Hockenhull J, Bagust A, et al. Systematic review and cost-effectiveness evaluation of ‘pill-in-the-pocket’ strategy for paroxysmal atrial fibrillation compared to episodic in-hospital treatment or continuous antiarrhythmic drug therapy. Health Technol Assess 2010;14(iii-iv):1-75.

563. Andrade JG, MacGillivray J, Macle L, et al. Clinical effectiveness of a systematic “pill-in-the-pocket” approach for the management of paroxysmal atrial fibrillation. Heart Rhythm 2018;15:9-16.

564. Jacoby JL, Cesta M, Heller MB, Salen P, Reed J. Synchronized emergency department cardioversion of atrial dysrhythmias saves time, money and resources. J Emerg Med 2005;28:27-30.

565. Xavier Scheuermeyer F, Grafstein E, Stenstrom R, et al. Thirty-day outcomes of emergency department patients undergoing electrical cardioversion for atrial fibrillation or flutter. Acad Emerg Med 2010;17:408-15.

566. Gronberg T, Nuotio I, Nikkinen M, et al. Arrhythmic complications after electrical cardioversion of acute atrial fibrillation: the FinCV study. Europace 2013;15:1432-5.

567. Bonfanti L, Annovi A, Sanchis-Gomar F, et al. Effectiveness and safety of electrical cardioversion for acute-onset atrial fibrillation in the emergency department: a real-world 10-year single center experience. Clin Exp Emerg Med 2019;6:64-9.

568. Bellone A, Etteri M, Vettorello M, et al. Cardioversion of acute atrial fibrillation in the emergency department: a prospective randomised trial. Emerg Med J 2012;29:188-91.

569. Ballard DW, Reed ME, Singh N, et al. Emergency department management of atrial fibrillation and flutter and patient quality of life at one month postvisit. Ann Emerg Med 2015;66:646-54.

570. Inacio JF, da Rosa Mdos S, Shah J, et al. Monophasic and biphasic shock for transthoracic conversion of atrial fibrillation: systematic review and network meta-analysis. Resuscitation 2016;100:66-75.

571. Kirkland S, Stiell I, AlShawabkeh T, et al. The efficacy of pad placement for electrical cardioversion of atrial fibrillation/flutter: a systematic review. Acad Emerg Med 2014;21:717-26.

572. Ramirez FD, Fiset SL, Cleland MJ, et al. Effect of applying force to selfadhesive electrodes on transthoracic impedance: implications for electrical cardioversion. Pacing Clin Electrophysiol 2016;39:1141-7.

573. Sadek MM, Chaugai V, Cleland MJ, et al. Association between transthoracic impedance and electrical cardioversion success with biphasic defibrillators: an analysis of 1055 shocks for atrial fibrillation and flutter. Clin Cardiol 2018;41:666-70.

574. Oral H, Souza JJ, Michaud GF, et al. Facilitating transthoracic cardioversion of atrial fibrillation with ibutilide pretreatment. N Engl J Med 1999;340:1849-54.

575. Wyse DG. Therapeutic considerations in applying rate control therapy for atrial fibrillation. J Cardiovasc Pharmacol 2008;52:11-7.

576. Scheuermeyer FX, Grafstein E, Stenstrom R, et al. Safety and efficiency of calcium channel blockers versus beta-blockers for rate control in patients with atrial fibrillation and no acute underlying medical illness. Acad Emerg Med 2013;20:222-30.

577. Segal JB, McNamara RL, Miller MR, et al. The evidence regarding the drugs used for ventricular rate control. J Fam Pract 2000;49:47-59.

578. Ahmad K, Dorian P. Rate control in atrial fibrillation: looking beyond the average heart rate. Curr Opin Cardiol 2006;21:88-93.

579. Groenveld HF, Crijns HJ, Van den Berg MP, et al. The effect of rate control on quality of life in patients with permanent atrial fibrillation: data from the RACE II (Rate Control Efficacy in Permanent Atrial Fibrillation II) study. J Am Coll Cardiol 2011;58:1795-803.

580. Ulimoen SR, Enger S, Pripp AH, et al. Calcium channel blockers improve exercise capacity and reduce N-terminal pro-B-type natriuretic peptide levels compared with beta-blockers in patients with permanent atrial fibrillation. Eur Heart J 2014;35:517-24.

581. Ulimoen SR, Enger S, Carlson J, et al. Comparison of four single-drug regimens on ventricular rate and arrhythmia-related symptoms in patients with permanent atrial fibrillation. Am J Cardiol 2013;111:225-30.

582. Aguilar M, Nattel S. Taking the pulse of atrial fibrillation: a practical approach to rate control. Can J Cardiol 2018;34:1526-30.

583. McAlister FA, Wiebe N, Ezekowitz JA, Leung AA, Armstrong PW. Meta-analysis: beta-blocker dose, heart rate reduction, and death in patients with heart failure. Ann Intern Med 2009;150:784-94.

584. Cullington D, Goode KM, Zhang J, Cleland JG, Clark AL. Is heart rate important for patients with heart failure in atrial fibrillation? JACC Heart Fail 2014;2:213-20.

585. Laskey WK, Alomari I, Cox M, et al. Heart rate at hospital discharge in patients with heart failure is associated with mortality and rehospitalization. J Am Heart Assoc 2015;4:e001626.

586. Kotecha D, Flather MD, Altman DG, et al. Heart rate and rhythm and the benefit of beta-blockers in patients with heart failure. J Am Coll Cardiol 2017;69:2885-96.

587. Cadrin-Tourigny J, Shohoudi A, Roy D, et al. Decreased mortality with beta-blockers in patients with heart failure and coexisting atrial fibrillation: an AF-CHF substudy. JACC Heart Fail 2017;5:99-106.

588. Head SJ, Milojevic M, Daemen J, et al. Mortality after coronary artery bypass grafting versus percutaneous coronary intervention with stenting for coronary artery disease: a pooled analysis of individual patient data. Lancet 2018;391:939-48.

589. Gjesdal K, Feyzi J, Olsson SB. Digitalis: a dangerous drug in atrial fibrillation? An analysis of the SPORTIF III and V data. Heart 2008;94:191-6.

590. Shah M, Avgil Tsadok M, Jackevicius CA, et al. Relation of digoxin use in atrial fibrillation and the risk of all-cause mortality in patients 65 years of age with versus without heart failure. Am J Cardiol 2014;114:401-6.

591. Eisen A, Ruff CT, Braunwald E, et al. Digoxin use and subsequent clinical outcomes in patients with atrial fibrillation with or without heart failure in the ENGAGE AF-TIMI 48 trial. J Am Heart Assoc 2017;6:e006035.

592. Lopes RD, Rordorf R, De Ferrari GM, et al. Digoxin and mortality in patients with atrial fibrillation. J Am Coll Cardiol 2018;71:1063-74.

593. Sethi NJ, Safi S, Feinberg J, et al. Digoxin versus placebo, no intervention, or other medical interventions for atrial fibrillation and atrial flutter: a protocol for a systematic review with meta-analysis and trial sequential analysis. Syst Rev 2017;6:71.

594. Ahmed A, Pitt B, Rahimtoola SH, et al. Effects of digoxin at low serum concentrations on mortality and hospitalization in heart failure: a propensity-matched study of the DIG trial. Int J Cardiol 2008;123:138-46.

595. Hofmann R, Wimmer G, Leisch F. Intravenous amiodarone bolus immediately controls heart rate in patients with atrial fibrillation accompanied by severe congestive heart failure. Heart 2000;84:635.

596. Tse HF, Lam YM, Lau CP, Cheung BM, Kumana CR. Comparison of digoxin versus low-dose amiodarone for ventricular rate control in patients with chronic atrial fibrillation. Clin Exp Pharmacol Physiol 2001;28:446-50.

597. Connolly SJ, Camm AJ, Halperin JL, et al. Dronedarone in high-risk permanent atrial fibrillation. N Engl J Med 2011;365:2268-76.

598. Van Gelder IC, Groenveld HF, Crijns HJ, et al. Lenient versus strict rate control in patients with atrial fibrillation. N Engl J Med 2010;362:1363-73.

599. Cooper HA, Bloomfield DA, Bush DE, et al. Relation between achieved heart rate and outcomes in patients with atrial fibrillation (from the Atrial Fibrillation Follow-up Investigation of Rhythm Management [AFFIRM] study). Am J Cardiol 2004;93:1247-53.

600. Wyse DG. Lenient versus strict rate control in atrial fibrillation some devils in the details. J Am Coll Cardiol 2011;58:950-2.

601. Steinberg BA, Kim S, Thomas L, et al. Increased heart rate is associated with higher mortality in patients with atrial fibrillation (AF): results from the Outcomes Registry for Better Informed Treatment of AF (ORBIT-AF). J Am Heart Assoc 2015;4:e002031.

602. Andrade JG, Roy D, Wyse DG, et al. Heart rate and adverse outcomes in patients with atrial fibrillation: a combined AFFIRM and AF-CHF substudy. Heart Rhythm 2016;13:54-61.

603. Lau DH, Thiyagarajah A, Willems S, et al. Device therapy for rate control: pacing, resynchronisation and AV node ablation. Heart Lung Circ 2017;26:934-40.

604. Kay GN, Ellenbogen KA, Giudici M, et al. The Ablate and Pace Trial: a prospective study of catheter ablation of the AV conduction system and permanent pacemaker implantation for treatment of atrial fibrillation. APT Investigators. J Interv Card Electrophysiol 1998;2:121-35.

605. Chatterjee NA, Upadhyay GA, Ellenbogen KA, et al. Atrioventricular nodal ablation in atrial fibrillation: a meta-analysis and systematic review. Circ Arrhythm Electrophysiol 2012;5:68-76.

606. Natale A, Zimerman L, Tomassoni G, et al. AV node ablation and pacemaker implantation after withdrawal of effective rate-control medications for chronic atrial fibrillation: effect on quality of life and exercise performance. Pacing Clin Electrophysiol 1999;22:1634-9.

607. Chatterjee NA, Upadhyay GA, Ellenbogen KA, Hayes DL, Singh JP. Atrioventricular nodal ablation in atrial fibrillation: a meta-analysis of biventricular vs right ventricular pacing mode. Eur J Heart Fail 2012;14:661-7.

608. Vijayaraman P, Subzposh FA, Naperkowski A. Atrioventricular node ablation and His bundle pacing. Europace 2017;19:iv10-6.

609. Yarlagadda B, Turagam MK, Dar T, et al. Safety and feasibility of leadless pacemaker in patients undergoing atrioventricular node ablation for atrial fibrillation. Heart Rhythm 2018;15:994-1000.

610. Okabe T, El-Chami MF, Lloyd MS, et al. Leadless pacemaker implantation and concurrent atrioventricular junction ablation in patients with atrial fibrillation. Pacing Clin Electrophysiol 2018;41:504-10.

611. Curtis AB, Worley SJ, Adamson PB, et al. Biventricular pacing for atrioventricular block and systolic dysfunction. N Engl J Med 2013;368:1585-93.

612. Dickstein K, Bogale N, Priori S, et al. The European cardiac resynchronization therapy survey. Eur Heart J 2009;30:2450-60.

613. Hayes DL, Boehmer JP, Day JD, et al. Cardiac resynchronization therapy and the relationship of percent biventricular pacing to symptoms and survival. Heart Rhythm 2011;8:1469-75.

614. Healey JS, Hohnloser SH, Exner DV, et al. Cardiac resynchronization therapy in patients with permanent atrial fibrillation: results from the Resynchronization for Ambulatory Heart Failure Trial (RAFT). Circ Heart Fail 2012;5:566-70.

615. Ganesan AN, Brooks AG, Roberts-Thomson KC, et al. Role of AV nodal ablation in cardiac resynchronization in patients with coexistent atrial fibrillation and heart failure a systematic review. J Am Coll Cardiol 2012;59:719-26.

616. Gasparini M, Kloppe A, Lunati M, et al. Atrioventricular junction ablation in patients with atrial fibrillation treated with cardiac resynchronization therapy: positive impact on ventricular arrhythmias, implantable cardioverter-defibrillator therapies and hospitalizations. Eur J Heart Fail 2018;20:1472-81.

617. Freemantle N, Lafuente-Lafuente C, Mitchell S, Eckert L, Reynolds M. Mixed treatment comparison of dronedarone, amiodarone, sotalol, flecainide, and propafenone, for the management of atrial fibrillation. Europace 2011;13:329-45.

618. Zimetbaum P. Antiarrhythmic drug therapy for atrial fibrillation. Circulation 2012;125:381-9.

619. Lafuente-Lafuente C, Valembois L, Bergmann JF, Belmin J. Antiarrhythmics for maintaining sinus rhythm after cardioversion of atrial fibrillation. Cochrane Database Syst Rev 2015;3:CD005049.

620. Hohnloser SH, Crijns HJ, van Eickels M, et al. Effect of dronedarone on cardiovascular events in atrial fibrillation. N Engl J Med 2009;360:668-78.

621. Connolly SJ, Crijns HJ, Torp-Pedersen C, et al. Analysis of stroke in ATHENA: a placebo-controlled, double-blind, parallel-arm trial to assess the efficacy of dronedarone 400 mg BID for the prevention of cardiovascular hospitalization or death from any cause in patients with atrial fibrillation/atrial flutter. Circulation 2009;120:1174-80.

622. Kirchhof P, Andresen D, Bosch R, et al. Short-term versus long-term antiarrhythmic drug treatment after cardioversion of atrial fibrillation (Flec-SL): a prospective, randomised, open-label, blinded end point assessment trial. Lancet 2012;380:238-46.

623. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med 1991;324:781-8.

624. Aliot E, Capucci A, Crijns HJ, Goette A, Tamargo J. Twenty-five years in the making: flecainide is safe and effective for the management of atrial fibrillation. Europace 2011;13:161-73.

625. Goldschlager N, Epstein AE, Naccarelli GV, et al. A practical guide for clinicians who treat patients with amiodarone: 2007. Heart Rhythm 2007;4:1250-9.

626. Wegener FT, Ehrlich JR, Hohnloser SH. Dronedarone: an emerging agent with rhythm- and rate-controlling effects. J Cardiovasc Electrophysiol 2006;17(suppl 2):S17-20.

627. Andrade JG, Champagne J, Dubuc M, et al. Cryoballoon or radiofrequency ablation for atrial fibrillation assessed by continuous monitoring: a randomized clinical trial. Circulation 2019;140:1779-88.

628. Stewart MT, Haines DE, Verma A, et al. Intracardiac pulsed field ablation: proof of feasibility in a chronic porcine model. Heart Rhythm 2019;16:754-64.

629. Haissaguerre M, Jais P, Shah DC, et al. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. N Engl J Med 1998;339:659-66.

630. Verma A, Jiang CY, Betts TR, et al. Approaches to catheter ablation for persistent atrial fibrillation. N Engl J Med 2015;372:1812-22.

631. Andrade JG, Macle L, Verma A, et al. Quality of life and health care utilization in the CIRCA-DOSE study. JACC Clin Electrophysiol 2020;6:935-44.

632. Deshmukh A, Patel NJ, Pant S, et al. In-hospital complications associated with catheter ablation of atrial fibrillation in the United States between 2000 and 2010: analysis of 93 801 procedures. Circulation 2013;128:2104-12.

633. Cheng EP, Liu CF, Yeo I, et al. Risk of mortality following catheter ablation of atrial fibrillation. J Am Coll Cardiol 2019;74:2254-64.

634. Khan SU, Rahman H, Talluri S, Kaluski E. The clinical benefits and mortality reduction associated with catheter ablation in subjects with atrial fibrillation: a systematic review and meta-analysis. JACC Clin Electrophysiol 2018;4:626-35.

635. Asad ZUA, Yousif A, Khan MS, Al-Khatib SM, Stavrakis S. Catheter ablation versus medical therapy for atrial fibrillation: a systematic review and meta-analysis of randomized controlled trials. Circ Arrhythm Electrophysiol 2019;12:e007414.

636. Blomstrom-Lundqvist C, Gizurarson S, Schwieler J, et al. Effect of Catheter Ablation vs Antiarrhythmic Medication on Quality of Life in Patients With Atrial Fibrillation: the CAPTAF randomized clinical trial. JAMA 2019;321:1059-68.

637. Bunch TJ, May HT, Bair TL, et al. Increasing time between first diagnosis of atrial fibrillation and catheter ablation adversely affects long-term outcomes. Heart Rhythm 2013;10:1257-62.

638. Wazni OM, Marrouche NF, Martin DO, et al. Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of symptomatic atrial fibrillation: a randomized trial. JAMA 2005;293:2634-40.

639. Cosedis Nielsen J, Johannessen A, Raatikainen P, et al. Radiofrequency ablation as initial therapy in paroxysmal atrial fibrillation. N Engl J Med 2012;367:1587-95.

640. Morillo CA, Verma A, Connolly SJ, et al. Radiofrequency ablation vs antiarrhythmic drugs as first-line treatment of paroxysmal atrial fibrillation (RAAFT-2): a randomized trial. JAMA 2014;311:692-700.

641. Andrade JG, Wells GA, Deyell MW, et al; for the EARLY-AF Investigators. Cryoablation or drug therapy for initial treatment of atrial fibrillation [epub ahead of print]. N Engl J Med, https://doi.org/10.1056/NEJMoa2029980.

642. Anselmino M, Matta M, D’Ascenzo F, et al. Catheter ablation of atrial fibrillation in patients with left ventricular systolic dysfunction: a systematic review and meta-analysis. Circ Arrhythm Electrophysiol 2014;7:1011-8.

643. Malhi N, Hawkins NM, Andrade JG, Krahn AD, Deyell MW. Catheter ablation of atrial fibrillation in heart failure with reduced ejection fraction. J Cardiovasc Electrophysiol 2018;29:1049-58.

644. Di Biase L, Mohanty P, Mohanty S, et al. Ablation Versus Amiodarone for Treatment of Persistent Atrial Fibrillation in Patients With Congestive Heart Failure and an Implanted Device: results from the AATAC multicenter randomized trial. Circulation 2016;133:1637-44.

645. Marrouche NF, Brachmann J, Andresen D, et al. Catheter ablation for atrial fibrillation with heart failure. N Engl J Med 2018;378:417-27.

646. Samuel M, Abrahamowicz M, Joza J, et al. Long-term effectiveness of catheter ablation in patients with atrial fibrillation and heart failure. Europace 2020;22:739-47.

647. Khaykin Y, Marrouche NF, Martin DO, et al. Pulmonary vein isolation for atrial fibrillation in patients with symptomatic sinus bradycardia or pauses. J Cardiovasc Electrophysiol 2004;15:784-9.

648. Natale A, Newby KH, Pisano E, et al. Prospective randomized comparison of antiarrhythmic therapy versus first-line radiofrequency ablation in patients with atrial flutter. J Am Coll Cardiol 2000;35:1898-904.

649. Spector P, Reynolds MR, Calkins H, et al. Meta-analysis of ablation of atrial flutter and supraventricular tachycardia. Am J Cardiol 2009;104:671-7.

650. Reithmann C, Hoffmann E, Spitzlberger G, et al. Catheter ablation of atrial flutter due to amiodarone therapy for paroxysmal atrial fibrillation. Eur Heart J 2000;21:565-72.

651. Budera P, Straka Z, Osmancik P, et al. Comparison of cardiac surgery with left atrial surgical ablation vs cardiac surgery without atrial ablation in patients with coronary and/or valvular heart disease plus atrial fibrillation: final results of the PRAGUE-12 randomized multicentre study. Eur Heart J 2012;33:2644-52.

652. Gillinov AM, Gelijns AC, Parides MK, et al. Surgical ablation of atrial fibrillation during mitral-valve surgery. N Engl J Med 2015;372:1399-409.

653. Phan K, Xie A, La Meir M, Black D, Yan TD. Surgical ablation for treatment of atrial fibrillation in cardiac surgery: a cumulative metaanalysis of randomised controlled trials. Heart 2014;100:722-30.

654. Jessurun ER, van Hemel NM, Defauw JJ, et al. A randomized study of combining maze surgery for atrial fibrillation with mitral valve surgery. J Cardiovasc Surg (Torino) 2003;44:9-18.

655. Gillinov AM, Bhavani S, Blackstone EH, et al. Surgery for permanent atrial fibrillation: impact of patient factors and lesion set. Ann Thorac Surg 2006;82:502-13 [discussion: 513-4].

656. von Oppell UO, Masani N, O’Callaghan P, et al. Mitral valve surgery plus concomitant atrial fibrillation ablation is superior to mitral valve surgery alone with an intensive rhythm control strategy. Eur J Cardiothorac Surg 2009;35:641-50.

657. Ad N, Damiano RJ Jr, Badhwar V, et al. Expert consensus guidelines: examining surgical ablation for atrial fibrillation. J Thorac Cardiovasc Surg 2017;153:1330-1354.e1331.

Back to top