Open access peer-reviewed chapter

Arrhythmias Post Coronary Artery Bypass Surgery

Written By

Bandar Al-Ghamdi

Submitted: 19 March 2017 Reviewed: 18 July 2017 Published: 20 December 2017

DOI: 10.5772/intechopen.70423

From the Edited Volume

Coronary Artery Bypass Graft Surgery

Edited by Wilbert S. Aronow

Chapter metrics overview

2,896 Chapter Downloads

View Full Metrics

Abstract

Arrhythmias are common after cardiac surgery such as coronary artery bypass grafting surgery. Although most of these arrhythmias are transient and have a benign course, it may represent a significant source of morbidity and mortality. Postoperative arrhythmias (POAs) include atrial tachyarrhythmias, ventricular arrhythmias, and bradyarrhythmias. The incidence of POAs has not changed despite improvements in anesthetic and surgical techniques. The tachyarrhythmias in the postoperative period include atrial fibrillation, atrial flutter, supraventricular tachycardia, and ventricular tachycardia. The clinical significance of each arrhythmia depends on several factors that include cardiac function, patient’s comorbidities, arrhythmia duration, and ventricular response rate. Tachycardia with uncontrolled ventricular rates can cause diastolic and later on systolic dysfunction, reduce cardiac output, and result in hypotension or myocardial ischemia. In the other hand, bradyarrhythmias may have a remarkable influence on patients with systolic or diastolic ventricular dysfunction. Arrhythmia management starts preoperatively with optimizing the patient’s condition and controlling patient’s risk factors, intra-operatively with careful attention to hemodynamic changes during surgery and uses appropriate anesthesia, and postoperatively with correction of temporary and correctable predisposing factors, as well as specific therapy for the arrhythmia itself. The POAs treatment urgency and management options are determined by the clinical presentation of the arrhythmia.

Keywords

  • coronary artery bypass surgery
  • arrhythmias
  • atrial fibrillation
  • ventricular arrhythmias
  • bradyarrhythmias

1. Introduction

Arrhythmias are common after cardiac surgery such as coronary artery bypass grafting (CABG) surgery and represent a significant source of morbidity and mortality. Although most of these arrhythmias are transient and have a benign course, it may prolong intensive care and hospital stay, and in rare instances, it may lead to mortality. Postoperative arrhythmias (POAs) include atrial tachyarrhythmias (ATs) and to a lesser extent ventricular arrhythmias (VAs) and bradyarrhythmias [1]. The incidence of POAs has not changed despite improvements in anesthetic and surgical techniques, and evidence suggests that its incidence may be increasing [2].

The clinical significance of each arrhythmia depends on several factors that include underlying cardiac function, patient’s comorbidities, arrhythmia duration, and ventricular response rate. So, POAs could be tolerated in some patients and a source of morbidity and mortality in others, depending on the interaction between these factors [1, 3]. Rapid ventricular rates with tachycardia can cause diastolic and later on systolic dysfunction, reduce cardiac output, and result in hypotension or myocardial ischemia [4, 5]. Bradydysrhythmias, particularly with the loss of atrial function, may have a remarkable influence on patients with systolic or diastolic ventricular dysfunction [6].

Arrhythmia management starts preoperatively with optimization of the patient’s condition and controlling patient’s risk factors. Intraoperatively, it includes careful attention to hemodynamic changes during surgery and uses appropriate anesthesia. Postoperatively, it includes correction of temporary and correctable predisposing factors, as well as specific therapy for the arrhythmia itself [7]. The POAs treatment urgency and management options are determined by the clinical presentation of the arrhythmia [7]. Self-terminating arrhythmias without overt cardiac disease often need no therapy. However, arrhythmias with hemodynamic instability, especially in patients with critical stress conditions like systemic infections or persistent pericardial effusion need urgent intervention to restore a stable clinical status [7].

The aim of this chapter is to review post-CABG arrhythmias pathophysiology and management.

Advertisement

2. Pathophysiology

The primary function of CABG is to reestablish perfusion to ischemic myocardium with utilizing autologous arteries and veins. This may be achieved by using different surgical techniques. The POAs pathophysiology, incidence, and clinical course may vary depending on the surgical techniques used. Initially, cardiac surgeries were performed on a beating heart, but with the development of cardiopulmonary bypass (CPB) machine and cardioplegia, most CABG surgeries were performed on a pump. However, interest in off-pump coronary artery bypass (OPCAB) surgery had revived in the 1990s [8]. Reported potential benefits of OPCAB include lower end-organ damage with less cerebrovascular accidents (CVA), fewer cognitive deficits, renal failure, less psychomotor defects, reduced systemic inflammation, and lower transfusion rates [9]. However, variable outcomes have been reported in studies comparing these strategies [9]. Minimally invasive surgery without use of CPB and through smaller incisions- and robotic-assisted approaches have also been developed [9]. This method is most often used for left internal mammary artery (LIMA) to left anterior descending artery (LAD) grafts. Additional benefits may also include reduced operative time, reduced recovery time, decreased the need for blood transfusion, less time under anesthesia, reduced duration of ICU stay, less pain, and an estimated 40% savings over conventional CABG [10].

The development of POAs is related to factors that influence the atrial and ventricular myocardium. These primarily include: a previous anatomic substrate, caused by degenerative changes typical of age and underlying disease, and electrical substrate derived from the perioperative processes that alter the membrane potentials, increase the dispersion of the refractory periods, and decrease the conduction velocity [11]. Inflammation and hyperadrenergic state appear to play a fundamental role in the development of postoperative tachyarrhythmias, favoring automatism [11]. Hypokalemia and hypomagnesemia characteristic of this period alter phase III of the membrane action potential, increasing the automatism, and slowing the conduction speed [11]. Atrial and ventricular ischemia due to hypoxemia is another contributing factor [11].

Several perioperative risk factors have been implicated in atrial and ventricular susceptibility to POAs, but their relative role is still uncertain. Risk factors for POAs may be classified into patient- and surgery-related factors [7].

2.1. Patient-related risk factors

Various patient-related risk factors have been described to cause POAs. These include:

2.1.1. Age

Increasing age is associated with age-related structural and electrophysiological changes that may lead to postoperative atrial tachyarrhythmias in the elderly. Old age has been demonstrated to be correlated with the development of POAs [2, 3, 1214].

2.1.2. Underlying structural heart disease

Patients with underlying structural heart disease are at higher risk of developing POAs compared to patients with a normal heart. Structural heart disease in the atria and ventricles provides a substrate for arrhythmia via abnormal automaticity, triggered activity, or reentry. Cardiac surgery patients often have the substrate of atrial enlargement and elevation of atrial pressures may function as a substrate for atrial arrhythmias. It is well known that large atrial size and fibrosis supports propagation of atrial reentrant circuits and helps in maintaining atrial fibrillation (AF). Similarly, patients with ventricular dysfunction, ventricular dilation, or fibrosis are at higher risk of having ventricular arrhythmias [4]. Other important risk factors for POAs include previous history of arrhythmias (e.g., AF), cardiac surgery, and POAs. Also, severe right coronary artery stenosis [15], sinus nodal or atrioventricular nodal branch disease [13, 16, 17], and mitral valvular disease (particularly rheumatic mitral stenosis) have been reported as risk factors for POAs. The preoperative brain natriuretic peptide plasma concentration is another predictor of POAs [18].

2.1.3. Other comorbidities

Noncardiac comorbidities have been reported to increase the risk of POAs especially AF. These include obesity [19], previous stroke, and history of chronic obstructive pulmonary disease [20].

2.2. Surgery-related risk factors

Cardiac surgery may lead to POAs via multiple surgery-related mechanisms and risk factors that include:

2.2.1. Trauma and inflammation

Cardiac surgery provokes a vigorous inflammatory response due to a variety of metabolic, endocrine, and immune changes known as the “stress response,” which has important clinical implications [21, 22] (Figure 1). Surgical trauma, blood loss or transfusion, hypothermia, and CPB are nonspecific activators of the inflammatory response [18, 19]. Surgical trauma may contribute to a higher degree of the inflammatory response compared to CPB [23]. These effects predispose to atrial and ventricular arrhythmias in the early postoperative period. Inflammatory mechanisms have been proposed for the development of postoperative AF (POAF) as its incidence peaks at early postoperative days. Inflammation may be related to the development of clinically aberrant or silent pericarditis. Unfortunately, clinical criteria, such as fever, pleuritic chest pain, pericardial rubs, and electrocardiogram changes correlate poorly with postoperative pericarditis and supraventricular arrhythmias [7]. However, patients with pericardial effusion in one study had a higher incidence of supraventricular arrhythmias (63% compared with 11% in patients without effusions) [24].

Figure 1.

Pathophysiologic changes in response to cardiopulmonary bypass and the extracorporeal circulation. ROS, reactive oxygen species; SIRS, systemic inflammatory response syndrome (from Ref. [22]).

2.2.2. Hemodynamic stress

Atrial and ventricular hemodynamic changes during CABG predispose to POAs. The risk factors for POAs include atrial changes at the time of cardiac surgery, such as acute atrial trauma from cannulation, enlargement, hypertension, and ischemia [7]. Postoperative pulmonary edema and postoperative pleural effusion requiring thoracentesis have also been described as possible risk factors [25]. Hemodynamic changes might trigger focal arrhythmias [7]. It is possible that atrial stretch, hypertension, pressure and volume shifts, and heightened catecholamine states can trigger AF foci from the pulmonary veins [26].

2.2.3. Ischemic injury

The coronary blood flow is interrupted during CABG surgery and CPB, and the heart is put under circulatory arrest. This interruption of coronary blood flow causes ischemia-reperfusion injury that is exacerbated by adverse neutrophil-mediated myocardial inflammation and injury [2729]. Atrial and ventricular ischemia or infarction triggers POAs [30]. Myocardial focal ischemia may occur due to endogenous or exogenous catecholamines, hypoxemia, hypercarbia, acid-base imbalances, drug effects, and mechanical factors. CPB, cross-clamp times, type of cardioplegia, and CABG surgical technique are also critical in determining ischemic injury. The incidence of AF has been demonstrated to be lower after OPCAB than conventional CABG. OPCAB is also associated with a lesser degree of inflammation [21].

2.2.4. Perioperative drugs

Beta-blocker withdrawal has been associated with an increased rate of postoperative supraventricular arrhythmias [31]. In contrary, some studies showed that preoperative digoxin use is a risk factor for POAs [2, 32], but not in the others [33]. Intravenous inotropic agents may be associated with POAs in some patients. The reported primary arrhythmias are sinus tachycardia (ST) and premature ventricular beats (PVCs), although other supraventricular (SVT) or ventricular arrhythmias (VT) have been reported. Clinically significant proarrhythmic effects with these agents appear to occur rarely. At conventional doses, intravenous inotropic agents are relatively safe concerning proarrhythmic effects. Inotropic agents increase sinoatrial node automaticity and decrease atrioventricular (AV) nodal conduction time [34, 35]. Dobutamine use has been reported to induce ventricular ectopic activity in 3–15% of patients [34]. Dopamine is more likely to be associated with a dose-related ST or AF [34]. Finally, short-term intravenous administration of the phosphodiesterase inhibitors amrinone and milrinone has been reported to cause PVCs and short runs of VT in up to 17% of patients [34]. Amiodarone and sotalol are useful and can be considered appropriate alternatives in high-risk patients [36]. Patients who need urgent CABG may benefit from intravenous and oral amiodarone combination in addition to beta-blockers. Although corticosteroids are associated with risk, it may be considered in selected CABG patients [36].

2.2.5. Electrolytes disorders

Hypokalemia leads to alteration of the electrophysiologic properties of cardiac myocytes with an increase in the action potential duration (increase in phase-3 depolarization), enhanced automaticity (increased slope of diastolic depolarization), and decreased conduction velocity [37]. These changes may provoke POAs [37]. Preoperative serum potassium levels of <3.5 mmol/L have a significant association with perioperative arrhythmias in patients undergoing elective CABG surgery [37].This association might be particularly evident in the atria, where changes in inward-rectifier potassium currents are supposed to act as profibrillatory mechanisms [38]. However, hypokalemia is more likely to be associated with VAs [38]. Moreover, it is worth noting that arrhythmogenesis is often multifactorial. Catecholamine release increases cellular potassium uptake and thus decreases serum potassium levels [39]. Serum potassium levels greater than 5.5 mmol/L appear to be associated with the development of POAF and atrial flutter (AFL) [37]. The role of magnesium remains controversial. The low serum magnesium levels—which is frequently seen after cardiac surgery—correlate with an increased incidence of POAs [7]. However, magnesium supplementation has produced conflicting results. Magnesium supplementation should be considered in all patients with hypomagnesemia [4041].

2.2.6. Other factors

The human epicardial fat pads (FPs) contain parasympathetic ganglia [42]. There are two posterior FPs with the first one located in the superior vena caval-atrial junction and contains postganglionic fibers that lead to the sinoatrial (SA) node. The second FP is located at the pulmonary vein-left atrium and contains postganglionic fibers that lead to the atrioventricular (AV) node [4345]. The anterior epicardial FP located in the aortopulmonary window that is routinely dissected and removed in CABG because it is located where the aortic cross-clamp is typically placed. Preservation of the human anterior epicardial FP during CABG decreases the incidence of POAF in one study [46], but not in another more recent study [47].

Advertisement

3. Postoperative atrial tachyarrhythmias (POATs)

3.1. Postoperative atrial fibrillation (POAF)

3.1.1. Epidemiology

AF is the most common complication seen after CABG surgery. The incidence of POAF is approximately 30% after isolated CABG, 40% after valve replacements or repair, and about 50% after combined CABG and valve surgeries [2, 4851]. The incidence of POAF increases with older age [2, 52, 53].

3.1.2. Diagnosis

The diagnosis of POAF is confirmed based on the telemetry and 12-lead electrocardiogram (ECG) recordings with an abrupt change in heart rate and rhythm, and loss of P waves [16, 54]. Atrial electrograms obtained from temporary atrial epicardial pacing wires that are often routinely placed at the time of cardiac surgery can be helpful in confirming the diagnosis of AF, AFL, and other forms of supraventricular tachycardia (SVTs) [54].

3.1.3. Clinical course

POAF usually occurs within 2–4 days after cardiac surgery, with a peak incidence on the second postoperative day [12, 55]. In POAF patients without a prior history of atrial arrhythmias, AF is usually self-limited. About 15–30% of POAF convert to sinus rhythm within 2 h and up to 80% within 24 h [56, 57]. The mean duration of AF in one report was 11–12 h [57], and >90% of the patients were in sinus rhythm 6–8 weeks after surgery [57, 58]. In another study, only 2 out of 112 patients who had paroxysmal AF after CABG were still in AF at 6 weeks [59].

3.1.4. Prognosis

Although POAF is often self-limiting, its clinical effects depend on ventricular rate, ventricular function, arrhythmia duration, symptoms, hemodynamic stability, and risk of thromboembolism. [60]. POAF is associated with increased postoperative thromboembolic risk and stroke [25, 6062]. In a series of 4507 patients, the incidence of stroke was significantly higher in those who developed POAF (3.3 versus 1.4%) [2]. Patient’s underlying comorbidities, such as older age, previous cerebrovascular disease (CVA), the presence of a carotid bruit, peripheral vascular disease (PVD), and CPB time, have an important role in the development of in-hospital stroke [6365]. In a review of 2972 patients undergoing CABG and/or valve surgery, POAF was associated with late onset stroke only if accompanied by a low cardiac output syndrome (3.9 versus 1.9%) [66]. Besides, POAF development is associated with a prolonged length of hospitalization [2, 25, 54]. The POAF is associated with an additional 2–4 days hospital stay after CABG surgery with an additional cost [54]. However, this effect seems to be less prominent with current cardiac surgical care [67]. Additionally, POAF may result in hemodynamic compromise [68], ventricular dysrhythmias [2], and iatrogenic complications associated with therapeutic interventions [53]. POAF may result in increased in-hospital and long-term mortality in a subset of patients [3, 60]. In a retrospective study of 6475 patients undergoing CABG at a single institution: 994 patients (15%) developed POAF. Higher in-hospital (7.4 versus 3.4%) and 4-year mortality (26 versus 13%) was noted in POAF patients but also with more comorbidities (i.e., older age, hypertension, and left ventricular hypertrophy) [60].

3.1.5. POAF management

The management of POAF should include the strategy for prevention and treatment of POAF when it develops. PAOF management starts with the optimization of medical comorbidities, if possible (e.g., hypoxia), and the correction of underlying electrolyte disturbances (e.g., potassium and magnesium abnormalities) [53]. POAF is treated similarly to AF in nonsurgical patients by rhythm control via pharmacological or electrical approach or heart rate control, and appropriate antithrombotic therapy.

3.1.5.1. Rhythm control versus heart rate control

In the atrial fibrillation follow-up investigation of rhythm management (AFFIRM) trial, the rate control versus rhythm control in nonsurgical patients with AF was studied and found that the use of rhythm control had no survival advantage, and it was associated with more frequent hospitalizations and adverse drug effects [69]. However, some studies involving patients with AF after cardiac surgery have suggested that rhythm control may offer advantages over rate control. This is still controversial and the evidence is inconclusive [11, 7072].

Treatment strategies of POAF aim to reduce symptoms, limit adverse hemodynamic effects, decrease the length of hospital stay, prevent readmissions, and improve survival [73]. The rhythm control strategy has the advantage of a rapid conversion to sinus rhythm, which restores atrial activity, functional capacity, and might reduce thromboembolic. The rate control strategy has the advantage of avoiding the potential adverse effects of antiarrhythmic drugs and complications associated with cardioversion [73]. In a recent trial, there was no difference in hospital admissions during a 60-day follow-up, with randomizing POAF patients to either rhythm control therapy with amiodarone or rate control [73]. As a result, the main aim of rhythm control therapy in POAF patients should be to improve AF-related symptoms. In asymptomatic patients and those with acceptable symptoms, rate control or deferred cardioversion preceded by anticoagulation is a reasonable approach [73].

In the following paragraph, rate control and rhythm control options will be discussed briefly.

(A) Rate control strategy: the rate control may be achieved by using beta-blockers, nondihydropyridine calcium channel blockers, digoxin, or a combination of these medications. Beta-blocker agents are the drug of choice, particularly for ischemic heart disease patients, because of the increased adrenergic stress in the postoperative period [53, 73, 74]. However, beta-blockers might be poorly tolerated or relatively contraindicated in patients with known bronchial asthma or bronchospastic lung diseases, active congestive heart failure, or AV conduction block [53]. The nondihydropyridine calcium channel blocker agents represent an alternative AV nodal blocking agent. Digoxin is less effective when the adrenergic tone is as high as in the postoperative period, but it may be used in patients with congestive heart failure. Amiodarone is another agent that can be used as it has been reported to be effective in controlling heart rate. Also, intravenous amiodarone administration has been associated with improved hemodynamic status [75, 76]. For further information about drugs used in AF rate control see Table 1.

Drug Route of admintration and doses Side effects sects Remarks
Beta-blockers
Atenolol Oral 25–100 mg QD Bradycardia, hypotension, fatigue, depression, negative inotropy, bronchospasm, AVB Decrease dose if CrCl <35
Bisoprolol Oral 2.5–10 mg QD As above Good choice for HF patients
Carvedilol Oral 3.125–25 mg BID As above Good choice for HF patients
Esmolol I.V. 500 mcg/kg bolus over 1 min, then 50–300 mcg/kg/min As above Only IV
Higher rate of hypotension
Metoprolol tartrate IV 2.5–5.0 mg bolus over 2 min; up to 3 doses
Oral 25–100 mg BID
As above
Metoprolol XL (succinate) Oral 50–400 mg QD As above Good choice for HF patients
Nadolol Oral 10 (usual initial adult dose 40 mg)–240 mg QD As above Dosage adjustments based on CrCl
Propranolol IV 1 mg over 1 min, up to 3 doses at 2-min intervals
Oral 10–40 mg up to 160–320 mg/day divided in BID to QID doses
80–160 to 320 mg QD (ER)
As above
Calcium channel blockers
Diltiazem IV 0.25 mg/kg bolus over 5 min followed by 0.05–0.15 mg/kg/h continuous infusion
Oral 30 mg TID/QID up to 480 mg/day
120–360 to 480 mg Q D (ER)
Bradycardia, hypotension, ankle swelling, exacerbation of HF, AVB Do not use in HF
Drug interaction via CYP3A4 including digoxin and warfarin
Verapamil IV 5–10 mg (0.075–0.15 mg/kg) over bolus at least 2 min; may give an additional 10 mg (0.15 mg/kg) after 30 min if no response, then 0.005 mg/kg/min infusion
Oral 80–120 mg TID up to 480 mg/day
180–480 mg QD or 240 BID (ER)
Bradycardia, AVB, hypotension, constipation, exacerbation of HF As diltiazem
Others
Digoxin IV 0.25 mg IV with repeat dosing to a maximum of 1.5 mg over 24 h
Oral 0.125–0.25 mg QD
Bradycardia, AVB, nausea, vomiting, visual disturbance Narrow therapeutic window
Adjust for renal failure
Drug interactions via p-glycoprotein
Amiodarone IV 150 mg over 10 min, followed by 1 mg/min continuous IV infusion for 6 h, then 0.5 mg/min continuous infusion for 18 h
Oral 400–800 mg/day PO in divided doses for 2–4 weeks to a total load of up to 10 g, then 100–200 mg QD
Bradycardia, hypotension, AV block, QTc prolongation, phlebitis on chronic use: Ocular, pulmonary, hepatic, hematological, neurological complications Monitor thyroid, liver and lung functions

Table 1.

Medications commonly used for atrial fibrillation rate control with its dosage and common possible side effects.

AVB, atrioventricular block; CrCl, creatinine clearance; BID, twice daily; h, hour; ER, extended release; HF, heart failure; IV, intravenous; mg milligram; min, minute; QD, once daily; QID, 4 times a day; QTc, correct QT interval; TID, 3 times a day.

(B) Rhythm control: the rhythm control could be archived by using a direct current cardioversion (electrical cardioversion) or antiarrhythmic drugs (pharmacological cardioversion). Electrical cardioversion is indicated on an urgent basis in hemodynamically unstable patients, acute heart failure, or myocardial ischemia. Also, it may be used electively to restore sinus rhythm when a pharmacologic attempt has failed to resume a sinus rhythm [53]. Rhythm control with antiarrhythmic medications is preferred in symptomatic patients despite rate control trial, or when the control of ventricular response is hard to achieve. Amiodarone [7779] or vernakalant [79, 80] have been efficient in converting POAF to sinus rhythm. Other antiarrhythmic medications that may be used include procainamide [80], ibutilide [81], and sotalol [82]. With ibutilide use, electrolyte imbalance should be corrected to avoid polymorphic ventricular tachycardia [82]. For further information about drugs used in AF rhythm control see Table 2.

Drug Usual doses Side effects sects Additional information
Vaughan Williams class IA
Disopyramide Oral
IR 100–200 mg q 6 h
ER 200–400 mg q 12 h
HF
Prolonged QT interval
Prostatism
Glaucoma
Metabolized by CYP3A4: caution with inhibitors (e.g., verapamil, diltiazem, ketoconazole, macrolide antibiotics, protease inhibitors, grapefruit juice) and inducers (e.g., phenytoin, phenobarbital, rifampin)
Avoid other QT interval prolonging drugs
Procainamide IV 15–17 mg/kg infused at a rate of 20–30 mg/min or alternatively100 mg IV every 5 min, max. 1 g
MD 1–4 mg/min
HF
Prolonged QT interval
May cause hypotension, myopathies, blood dyscrasias, and SLE-like syndrome
Drug of choice for WPW with AF
Avoid other QT interval prolonging drugs
Adjust for renal failure
Quinidine Oral
IR 200–300 mg q 6–8 h up to 600 mg q 6 h
ER 324 mg–648 mg q 8–12 h
Prolonged QT interval
Diarrhea
Bradycardia, AV block, bundle-branch block, digitalis toxicity
Inhibits CYP2D6: ⇧ concentrations of metoprolol, tricyclic antidepressants, antipsychotics; ⇩ efficacy of codeine
Inhibits P-glycoprotein:⇧ digoxin concentration
Vaughan Williams class IC
Flecainide IV 1.5–3 mg/kg over 10-20 min
Oral
LD 200 (wt < 70 kg)–300 mg (wt > 70 kg),
MD 50–200 mg BID max. 400 mg/day
Sinus or AV node dysfunction
HF
CAD
Atrial flutter
Brugada syndrome
Renal or liver disease
May cause blurred vision
Metabolized by CYP2D6 (inhibitors include quinidine, fluoxetine, tricyclics; also genetically absent in 7–10% of population) and renal excretion (dual impairment can ⇧⇧ plasma concentration)
Decrease dose if CrCl < 35
Propafenone IV 1.5–2 mg/kg over 10–20 min
Oral
IR: 150–300 mg q 8 h
ER: 225–425 mg q 12 h
(Oral LD 450 mg (wt < 70 kg)–600 mg (wt > 70 kg), MD 450–900 mg/d divided into q 8 h (IR), or 12 h (ER))
Sinus or AV node dysfunction or Infranodal conduction disease
HF
CAD
Atrial flutter
Brugada syndrome
Liver disease
Asthma may cause dysgeusia
Metabolized by CYP2D6 (inhibitors include quinidine, fluoxetine, tricyclics; also genetically absent in 7–10% of population)—poor metabolizers have ⇧beta blockade
Inhibits P-glycoprotein: ⇧digoxin concentration
Inhibits CYP2C9: ⇧warfarin concentration (⇧INR 25%)
Decrease dose in hepatic failure
Vaughan Williams class III
Amiodarone IV LD 150 mg over 10 min; followed by 1 mg/min for 6 h; then 0.5 mg/min for 18 h or change to oral dosing; after 24 h, consider decreasing dose to 0.25 mg/min
Oral 400–600 mg daily in divided doses for 2–4 wk; maintenance typically 100–200 mg QD
Sinus or AV node dysfunction
Infranodal conduction disease
Lung disease
Prolonged QT interval
Inhibits most CYPs to cause drug interaction: ⇧concentrations of warfarin (⇧INR between 0–200%), statins, many other drugs
Inhibits P-glycoprotein: ⇧digoxin concentration
Dronedarone Oral 400 mg BID Bradycardia
HF
Liver disease
Thyriod disease
pulmonary fibrosis
Prolonged QT interval
Metabolized by CYP3A: caution with inhibitors (e.g., verapamil, diltiazem, ketoconazole, macrolide antibiotics, protease inhibitors, grapefruit juice) and inducers (e.g., phenytoin, phenobarbital, rifampin) Inhibits CYP3A, CYP2D6, P-glycoprotein: ⇧concentrations of some statins, digoxin, beta blockers, sirolimus, tacrolimus
Avoid in long-standing persistent or permanent AF and HF
Dofetilide Oral 125–500 mcg BID if CrCl >60, 250 mcg if CrCl 40–60, 125 mcg if CrCl 20–40
Decrease MD if QTc increased by >15% of >500 ms 2–3 h after dose or consider discontinuing it
Prolonged QT interval and torsades de pointes
Renal disease
Hypokalemia hypomagnesaemia
AV block, bradycardia, sick sinus syndrome
Adjust dose for renal function, body size, and age (avoid if CrCl < 20)
Drug interactions via CYP3A4: CI to use with verapamil, cimetidine, ketoconazole, trimethoprim, prochlorperazine, HCTZ, and megestrol
Discontinue amiodarone at least 3 m before initiation
Avoid other QT interval prolonging drugs
Ibutilide IV 1 mg over 10 min; may repeat 1 mg
once if necessary (if weight <60 kg, use 0.01 mg/kg)
Prolonged QT interval and torsade de pointes hypotension
CAD
HF
Mointor K and mg level
Sotalol Oral 40–160 mg q12 h
IV 75–150 mg QD or BID over 5 h (only if patient cannot take oral)
Prolonged QT interval
Sinus or AV nodal dysfunction
HF
Renal excretion: CI if Cr Cl <40 decrease dose if CrCl 40–60
Risk of torsade de pointes ( do not initiate sotalol therapy if the baseline QTc is longer than 450 ms. If the QT interval prolongs to 500 ms or greater, the dose must be reduced, the duration of the infusion prolonged or the drug discontinued) Avoid other QT interval prolonging drugs correct hypokalemia/hypomagnesemia

Table 2.

Medications commonly used for atrial fibrillation Rythm Control with its dosage and major pharmacokinetic and drug interactions.

ACC AF [74]; JACC [53]. AF, atrial fibrillation; AV, atrioventricular; BID, twice daily; CAD, coronary artery disease; CI, contraindicated; CrCl, creatinine clearance; ER, extended release; h, hour; HCTZ, hydrochlorothiazide; HF, heart failure; IL, immediate release; IV, intravenous; LD, loading dose; INR, international normalized ratio; MD, maintenance dose; min, minute; max, maximum; SLE, systemic lupus erythematosus; Q, every; QD, once daily; wt, weight. http://www.pdr.net/

3.1.5.2. Anticoagulation

POAF is associated with poor short- and long-term outcomes, including high rates of early and late stroke, and late mortality as mentioned earlier. However, the indication and timing of anticoagulation in POAF patients should take into consideration the risk of postoperative bleeding. Oral anticoagulation at discharge has been associated with a reduced long-term mortality in patients with POAF [83] but without evidence from controlled trials [75]. POAF that persists for longer than 48 h should be anticoagulated with warfarin or nonvitamin K antagonist oral anticoagulants (NOACs). The NOACs are available for the treatment of nonvalvular AF. NOACs have been found to be as efficacious or even superior to warfarin in the prevention of stroke in nonvalvular AF patients with high risk of thromboembolism, with similar to lower rates of major bleeding, and also lower rates of intracranial hemorrhage [84].

3.1.6. Prevention of POAF

Beta-blockers are effective in reducing POAF and SVTs. Propranolol uses reduced POAF incidence from 31.7% in the control group to 16.3% in the treatment group [85]. In the majority of beta-blocker studies, it is administered postoperatively [86]. Amiodarone reduced the incidence of POAF and hospital stay compared to beta-blocker therapy in several meta-analyses [8689]. Prophylactic administration of sotalol may be considered for patients at risk of developing AF after cardiac surgery [76, 90, 91]. Also, administration of colchicine postoperatively may reduce POAF [75, 92]. Statin use preoperatively did not prevent POAF in a prospective controlled trial [93], despite that initial reports from meta-analyses were encouraging [9496].

Other therapies for the prevention of POAF have been studied in small trials, but have not demonstrated clear beneficial effects [76]. These include angiotensin converting enzyme inhibitors (ACEIs) [97], magnesium [85, 98, 99], n-3 polyunsaturated fatty acids [100108], corticosteroids [109111], and posterior pericardiectomy [112]. Conflicting results have also been reported for acetylcysteine [113], and sodium nitroprusside [114].

Nonpharmacologic therapy with atrial pacing has been tested in various studies [7]. One meta-analysis showed a significant reduction in POAF with atrial pacing (OR 0.57, 95% CI 0.38–0.84) [67], and most [115117] but not all [118, 119] published studies showed benefit with this therapy. Besides, there are conflicting findings as to the relative value of the different types of atrial pacing [115, 116].

3.2. Postoperative atrial flutter (POAFL)

Unlike POAF, POAFL after CABG is not well studied. In a single-center study with 80 consecutive patients who underwent CABG with no previous history of AFL, 16 patients (20%) had documented POAFL. Ten of these patients showed temporary AFLs that were curable without radiofrequency catheter ablation (RFCA), and 37.5% of the patients with POAFL (i.e., 7.5% of the patients after CABG) showed sustained or repeated AFL with subjective symptoms [120]. In another study that looked at ATs late after open heart surgery, it was found that cavotricuspid isthmus (CTI)-dependent AFL was the most common. Atypical AFL becomes progressively more widespread with more extensive atriotomy [121]. AFL and ATs that developed late after cardiac surgery are believed to be due to scars created by incisions applied to the right and/or left atrium either for establishing extracorporeal circulation or access to intracardiac structures (coronary sinus, interatrial or interventricular septum, atrioventricular valves, etc.) [122]. The scars created by these incisions play a significant role in the development of ATs, months or years after surgery [123, 124].

AFL in the early postoperative period is managed as POAF with rate control or rhythm control and anticoagulation based on arrhythmia duration and patient risk factors. On long-term catheter ablation of AFL is an effective, safe, and potentially curative procedure.

3.3. Supraventricular tachycardia (SVT)

3.3.1. Epidemiology

Sinus tachycardia (ST) represents an appropriate autonomic response to a physiological stress. The upper limit of normal rate for sinus tachycardia is calculated from the formula (220 bpm minus age) [125]. Inappropriate ST may be seen in some patients, especially with young age, but it is rare and should be considered a diagnosis of exclusion [125]. The term ‘SVT’ refers to paroxysmal tachyarrhythmias that require atrial or AV nodal tissue, or both, for their initiation and maintenance [126]. It is typical of a sudden or paroxysmal onset and includes AV nodal reentrant tachycardias (AVNRT), AV reentrant tachycardias (AVRT), and atrial tachycardias. The overall incidence of perioperative arrhythmias in noncardiac surgery varies from 16 to 62% with intermittent ECG monitoring and up to 89% with continuous Holter monitoring [127]. It is more likely to be supraventricular than ventricular in origin [127]. In small study, the incidence of persistent SVT in noncardiac surgery patients was 2% during surgery and 6% in the postoperative period [128].

3.3.2. Diagnosis

12-lead ECG and rhythm strips during tachycardia are diagnostic and may give an impression about the most likely diagnosis. Although ST is usually easy to diagnose on 12-lead ECG, the presence of first-degree AV block, which is not uncommon after cardiac surgery, may give ECG appearance that mimics SVT due to P wave merge with T wave (P wave hidden within T wave). ECG features of ATs including SVTs are shown in Table 3.

Table 3.

Differential diagnosis of atrial tachyarrhythmias.

3.3.3. Clinical course

ATs occur most frequently 2–3 days postsurgery and are likely related to sympathetic stimulation associated with an inflammatory response [129]. Patients with known SVT may have an exacerbation of their tachycardia in the perioperative period. However, SVT may be diagnosed for the first time in the perioperative period [2, 7, 130, 131]. SVT is often associated with a high sympathetic tone, but other precipitants may contribute to its occurrence. The clinical symptoms, time of onset, and natural course of ATs are identical in patients with cardiac, thoracic, or other surgery.

3.3.4. Prognosis

The prognosis of perioperative SVTs is good, but it may be associated with increased hospital stay [128].

3.3.5. Management

The SVT management, in general, depends on the hemodynamic status of the patient. If the patient with SVT is hemodynamically unstable, synchronized cardioversion is recommended for acute termination of the tachycardia when vagal maneuvers or adenosine is ineffective or not feasible [132]. Before initiating specific drug therapy for acute SVT in hemodynamically stable patients, it is important to assess and correct possible precipitating factors such as respiratory failure or electrolyte imbalance. SVT may respond to vagal maneuver if the patient can do it. Adenosine might be used if there is no contraindication. SVT also responds to rate control drugs such as beta-blockers (e.g., esmolol, metoprolol, bisoprolol) or nondihydropyridine calcium channel antagonists (e.g., diltiazem, verapamil). Intravenous (IV) digoxin, IV amiodarone, adenosine, IV or oral beta-blockers, diltiazem, and verapamil are potentially harmful in acute treatment in patients with pre-excited AF (AF in patients with Wolff-Parkinson-White (WPW) syndrome) [133]. Of note, atrial tachycardia unifocal or multifocal usually respond to rate control drugs but are not amenable to direct current cardioversion. Cardiac electrophysiology study with catheter ablation is an effective long-term management for recurrent SVT.

Advertisement

4. Postoperative ventricular tachyarrhythmias (POVTAs)

The postoperative ventricular tachyarrhythmias (POVTAs) range from isolated PVC to VT or ventricular fibrillation (VF).

4.1. Premature ventricular complexes (PVCs)

4.1.1. Epidemiology

Isolated PVCs including nonsustained ventricular tachycardia (NSVT) are seen in about 50% of patients during and after cardiac surgery [134]. PVCs can be related to electrolyte or other metabolic imbalances [7].

4.1.2. Diagnosis

PVCs may be seen on continuous telemetric monitoring and 12-lead ECG, however, careful evaluation of the ECG tracing is needed to be distinguished from atrial ectopy with aberrant ventricular conduction [7].

4.1.3. Clinical course

Patients with postoperative PVCs may be asymptomatic or may have palpitations with a skipped beat, or dizziness. It is rarely associated with hemodynamic instability.

4.1.4. Prognosis

Patients with isolated and noncomplicated PVCs postoperatively do not exhibit increased risk of malignant VAs [135, 136]. On the contrary, frequent PVCs (>30 per hour) may reduce ventricular function and therefore have an adverse impact on the short-term outcome. There was no significant difference in mortality in patients with versus patients without frequent postoperative PVCs and NSVT (8 versus 5%), at an average follow-up of 3 years, in a study including 185 postoperative patients [137]. However, in another study of 126 patients with postoperative PVCs, it was shown that patients with left ventricular ejection fraction (LVEF) of <40% had a 75% mortality rate and 33% incidence of sudden death at an average follow-up of 15 months, whereas none of the patients with preserved left ventricular function had sudden death [135]. Thus, PVCs are not related to mortality with good LV function, and long-term outcome after cardiac surgery seems to be closely related to the left ventricular function.

4.1.5. Management

Correction of any reversible cause of ventricular arrhythmias should be performed. Hemodynamically stable and asymptomatic PVCs do not usually need treatment with antiarrhythmic therapy on short or long-term. Lidocaine has been used with a successful result in reducing hemodynamically significant or symptomatic PVCs, but without improving mortality. Empirical use of class I antiarrhythmic drugs for suppression of frequent and/or complex PVCs had no beneficial effects on mortality rate and may be harmful as shown in several studies in another setting [138, 139]. Additionally, overdrive pacing, using either atrial or atrioventricular sequential pacing, has been used without significant results [138, 139]. Patients with asymptomatic NSVT after cardiac surgery and preserved LVEF generally have a favorable long-term prognosis and do not require invasive workup with an electrophysiology study. The use of implantable cardioverter defibrillators (ICDs) has shown no benefits in improving prognosis in this population [140].

4.2. Ventricular tachyarrhythmias

4.2.1. Epidemiology

Sustained VT and VF rarely occur after cardiac surgery with an incidence of 0.4–1.7% in most of the studies [138, 141], but an incidence of 3.1% has been reported [142]. Furthermore, it is life threatening and affects outcome [134, 143].

4.2.2. Pathophysiology and risk factors

Pathophysiology of POAs, in general, was disused in item 2. Coronary artery disease (CAD) leads to a broad spectrum of changes and may trigger arrhythmia mechanisms via enhanced automaticity, triggered activity, and reentry. While myocardial infarction (MI) related scar constitutes the clinical model of reentry [144], focal activation due to abnormal automaticity is the primary mechanism involved in the VT during acute ischemia [145]. Early and delayed after depolarization result from focal discharge by calcium overload and triggered activity is another likely mechanism of VT initiation in ischemia, but this needs to be proven experimentally thus far [146, 147]. Acute ischemia activates the adenosine triphosphate-sensitive potassium (K-ATP) channels, causing an increase in extracellular potassium along with acidosis and hypoxia in the cardiac muscle. As a result of the minor increases in extracellular potassium depolarize the myocardiocyte’s resting membrane potential, which can increase tissue excitability in early phases of ischemia [145]. The mechanism underlying the VT associated with healed or healing MI is reentry in more than 95% of cases [144].

Complex ventricular arrhythmias (VAs) are associated with multiple risk factors [7]. Based on clinical studies, the conditions associated with VAs after cardiac surgery may include: increased age, female gender, presence of unstable angina, congestive cardiac failure, hemodynamic instability, preoperative use of inotropes, preoperative use of IABP, emergency surgery, electrolyte disturbances, hypoxia, hypovolemia, myocardial ischemia/infarction, acute graft closure, reperfusion after cessation of CPB, and inotropes antiarrhythmic drugs use, on-pump surgery, and PVD [134, 135, 141, 143, 148].

4.2.3. Diagnosis

In addition to a clinical history and a physical exam, the general evaluation of a patient with CAD and suspected or documented VAs includes performing a 12-lead ECG and an echocardiogram. Telemetry monitoring with careful evaluation of VAs initiation and termination is very helpful. Based on ECG criteria, wide complex tachycardias (WCT) may be either ventricular or SVT with aberrancy. However, in patients with structural heart disease like prior infarction, the diagnosis is mostly VT. If feasible, a 12-lead ECG and atrial electrograms through temporary epicardial wires placed at the time of cardiac surgery should be obtained. The presence of AV dissociation strongly suggests VT [138]. Although the ECG diagnosis of a WCT is challenging, it is important to remember that VT is the cause in at least 80% of cases [149].

4.2.4. Clinical course

Clinical presentation of patients post cardiac surgery with VTs is variable. The hemodynamic state of these patients depends mainly on the rate of the tachyarrhythmia and the left ventricular function. Therefore, some patients may be asymptomatic. Other patients with VT may complain of palpitations, dyspnea, or chest discomfort as their main symptoms. VTs may present with syncope and sudden cardiac death as a result of hemodynamic compromise. Incessant VT, even if it is hemodynamically stable, can lead to hemodynamic deterioration and heart failure [150, 151].

4.2.5. Prognosis

The prognosis is correlated with the type of arrhythmia and the type and degree of structural heart disease [7]. As mentioned earlier, PVCs and NSVT generally have no impact on the outcome. However, patients with sustained VAs have poorer short- and long-term prognosis.

POVAs predicts higher in-hospital mortality (21.7–31.5%) compared with (1.4–2.9%) in control [134, 143, 148]. In one study, POVAs was associated with increased long-term mortality over a mean follow-up of 3.5 years. Patients with POVAs had a high risk of death in the POVAs group during the first 6 postsurgical months (6-month survival of POVAs 59.8 versus 93.8% for POVAs free group). This difference in survival persisted over time [148].

4.2.6. Management of POVAs

Asymptomatic PVCs and hemodynamically stable short runs of NSVT do not need specific intervention, and the correction of any reversible cause of VAs is generally sufficient. Postoperative sustained VAs treatment follows the ICD indications used in other clinical settings [150, 152]. However, the postoperative patients require close attention to the identification and treatment of reversible causes of arrhythmia like electrolyte or other metabolic disturbances, myocardial ischemia, or mechanical complications of surgery [7]. Sustained VAs should be promptly cardioverted either by drugs infusion or electrically based on hemodynamic stability. Hemodynamically stable sustained VTs may be initially treated with antiarrhythmic drugs infusion.

4.2.6.1. Pharmacological treatment

It includes antiarrhythmic medication use and standard medical therapy. The most commonly used antiarrhythmic agents include:

- Amiodarone: IV amiodarone is frequently used as a first-line treatment for VAs as it is better tolerated in patients with low ejection fraction than the other antiarrhythmic drugs. The recommended starting dose of Cordarone I.V. is 1000 mg over the first 24 h of therapy. It is usually delivered by bolus infusion of 150 mg over 10–15 min, followed by 1 mg/min for 6 h, then 0.5 mg/min infusion for 18 h. The alternative dose would be 300 mg over 1 h then infusion at 50 mg/h. Additional 150 mg blouses may be given but frequent boluses during the first 24 h should be limited due to the risk of hepatic toxicity [153].

- Lidocaine: it is generally a good choice if ischemia is suspected. Lidocaine is administered as a bolus of 0.75–1.5 mg/kg, followed by an IV infusion of 1–4 mg/min (the maximal dose is 3 mg/kg/h). In elderly patient and patients with congestive heart failure or hepatic dysfunction, the lidocaine dose should be reduced [153].

- Procainamide: it is often a second line drug, and it is given as loading dose of 15–18 mg/kg administered as a slow infusion over 25–30 min or 100 mg/dose. The infusion rate should not exceed 50 mg/min. The loading dose may be repeated every 5 min as needed to a total dose of 1 g. However, it should be stopped if hypotension occurs, or QRS complex widens by 50% of its original width. This is followed by a maintenance dose of 1–4 mg/min by continuous infusion. The procainamide maintenance infusion should be reduced by 1/3 in patients with moderate renal or cardiac impairment and by 2/3 in patients with severe renal or cardiac impairment [153].

Standard medical therapy includes beta-blockers, and ACEIs drugs have been demonstrated to improve long-term survival particularly in patients with left ventricular dysfunction.

4.2.6.2. Nonpharmacological management

- Overdrive pacing: in patients with slower VTs who have ventricular epicardial wires, overdrive pacing may be performed. Electrical cardioversion/defibrillation should be easily available because of the possibility of acceleration of the VT or degeneration to VF [154].

- Electrical cardioversion/defibrillation: in patients with cardiac arrest, basic life support (BLS) and advanced cardiovascular life support (ACLS) should be followed. Electrical defibrillation should be performed for VF, pulseless VT, and hemodynamically unstable VT. Electrical cardioversion may be used for stable sustained VT as the first choice or for those who do not respond to antiarrhythmic medications. The recommended energy with current biphasic defibrillators ranges from 150 to 200 Joules. Sedation with short-acting agents should precede cardioversion in awake patients [154].

- Emergency mechanical support: in postoperative patients who are not responding to standard resuscitation maneuvers, initiation of emergency CPB in the intensive care unit may be considered. In one study, CPB use in a postoperative cardiac arrest was associated with a 56% long-term survival rate with a 22% incidence of soft tissue infections and no mediastinitis [154].

- Implantable cardioverter-defibrillator (ICD) therapy:

In the absence of a reversible cause of sustained VT or cardiac arrest after CABG, long-term management may include electrophysiology study and eventually an ICD implantation. Patients with NSVT, prior MI, and left ventricular dysfunction (LVEF <40%) may be considered for electrophysiology testing and implantation of an ICD if a sustained ventricular arrhythmia is induced [152, 155]. Multicenter automatic defibrillator implantation trial (MADIT) study [152] excluded subjects within 2 months after CABG and 3 months after percutaneous transluminal coronary angioplasty (PTCA), and MADIT-II study [156] excluded subjects within 3 months after revascularization. Conversely, early revascularization was permitted in MUSTT (Multicenter Unsustained Tachycardia Trial) study [155], which enrolled subjects at least 4 days after revascularization, and sudden cardiac death in heart failure trial (SCD-HeFT) study [157] made no specific exclusion on the timing of revascularization. However, in SCD-HeFT, the median time from CABG to enrollment was 3.1 years, and from PCI to enrollment was 2.3 years. Therefore, ICD implantation within 90 days of coronary revascularization for patients who otherwise meet ICD implant criteria for primary prevention of sudden cardiac death (SCD) is not addressed in the published device-based therapy guidelines. Revascularization has significant time-dependent benefits. In fact, MADIT-II study showed that the efficacy of ICD therapy in patients with ischemic left ventricular dysfunction is time dependent, with a significant life-saving benefit in patients receiving device implantation more than 6 months after coronary revascularization (CR). The lack of ICD benefit early after CR may be related to a relatively small risk of SCD during this period [158]. Although, sudden cardiac arrest (SCA) has a higher incidence in patients with reduced LVEF in the months after acute MI and/or following revascularization [159, 160]. The two randomized controlled trials, defibrillator in acute myocardial infarction (DINAMIT) and immediate risk stratification improves survival (IRIS), showed that early ICD implantation does not reduce mortality [161, 162]. In both of those trials, there was a reduction in arrhythmic death, which was counteracted by a concomitant increase in death due to other causes [163]. Similarly, the coronary artery bypass graft (CABG)-patch trial [164] examined ICD implantation at the time of elective CABG surgery showed a small decrease in arrhythmic death, but no benefit for overall mortality in patients with preoperative LVEF ≤35%. However, one should keep in mind that the epicardial ICDs tested in this trial differed significantly from the current transvenous endocardial ICD systems. A retrospective study evaluating ICD implantation within 3 months of cardiac surgery suggested the benefit of ICD implantation for secondary prevention. In this study, 164 patients were with ICD implantation within 3 months of cardiac surgery; 93 of these patients had an ICD for sustained pre or postoperative VT or VF requiring resuscitation. During the mean follow-up of 49 months; the primary endpoint was total mortality (TM) and/or appropriate ICD therapy (ICD-T), and secondary endpoints are the TM and ICD-T, and individual end points of TM and ICD-T were observed in 52 (56%), 35 (38%), and 28 (30%) patients, respectively, with 55% of TM, and 23% of ICD-T occurring within 2 years of implant [165].

Overall, ICD for primary prevention of SCD can be useful in patients who are within 90 days of revascularization and who are not within 40 days of an acute MI if:

- they are previously qualified for primary prevention of SCD or

- revascularization is unlikely to result in an improvement in LVEF to level >35% [166].

ICD for secondary prevention (i.e., in patients resuscitated from cardiac arrest due to VT/VF) is recommended for patients within 90 days of revascularization who have:

- previously satisfied criteria for ICD implantation if they have abnormal left ventricular function or

- SCD is unlikely related to myocardial ischemia/injury and have normal left ventricular function [166].

ICD implantation can be useful in patients who are within 90 days of revascularization if SCD was not related to acute myocardial ischemia/injury and who were subsequently found to have coronary artery disease that is revascularized with normal left ventricular function, or SCD was not related to acute myocardial ischemia/injury and who were subsequently found to have coronary artery disease that is revascularized with normal left ventricular function [166]. ICD is not recommended in patients within 90 days of revascularization who were resuscitated from cardiac arrest due to VT that was related to acute MI/injury, with normal left ventricular function, and who undergo complete coronary revascularization [166]. ICD with appropriately selected pacing capabilities is recommended in patients within 90 days of revascularization who require nonelective permanent pacing, who would also meet primary prevention criteria for implantation of an ICD, and in whom recovery of left ventricular function is uncertain or not expected [166].

An alternative approach for primary prevention of SCD in patients with ischemic cardiomyopathy and low LVEF undergoing revascularization would be the use the wearable cardioverter-defibrillator vest during the 3 months waiting period after revascularization until LVEF is reassessed and design made about permanent ICD implantation [163].

- Ventricular tachycardia ablation:

There are no studies of VT ablation in POVAs situation. In patients with extensive structural abnormalities, especially those with prior MI, multiple morphologies of VT might develop. Therefore, VT ablation does not eliminate the need for ICD and/or antiarrhythmic therapy. VT episodes might occur in up 0–60% of patients who have received an ICD for secondary prevention and in 2.5–12% of patients with ICD implanted for primary prevention [167]. Because antiarrhythmic drugs do not eliminate the risk of VAs, VT catheter ablation may be needed to reduce the frequency of VT episodes, especially patients with incessant VT or frequent ICD therapy [149]. Ablation is usually indicated in cases of recurrent, monomorphic VT arising from a specific substrate that can be targeted by mapping techniques.

Advertisement

5. Postoperative bradyarrhythmias (POBAs)

5.1. Epidemiology

Bradyarrhythmias (BAs) are common after cardiac surgery, but it mostly consists of transient episodes of low ventricular heart rate. The conduction defects post cardiac surgery include sinus node dysfunction, partial and complete bundle branch blocks, and various degrees of atrioventricular (AV) block. The right bundle branch block (RBBB) was the most frequently noted abnormality [168]. Bradyarrhythmias may decrease cardiac output in patients with relatively fixed stroke volumes. The risk of developing conduction disturbances after CABG or valvular surgery leading to permanent pacemaker (PPM) implantation is about 0.4–1.1% of patients after isolated CABG and 3–6% after heart valve operations [169171]. It seems that in the current surgical era that the incidence of postoperative PPM implantation has decreased due to improvements in surgical techniques, technological innovations and enhanced understanding of the mechanisms of injury [172]. However, some studies have shown an increased incidence of PPM implantation after cardiac surgery after the year 2000 [173].

5.2. Pathophysiology and risk factors

Conduction disorders after cardiac surgery are explained by one of the following two mechanisms: (1) direct trauma to the conduction system in operative procedures in proximity to the sinoatrial or AV nodes or the His bundle; or (2) ischemic injury to the conduction system due to extensive coronary artery disease might compromise myocardial protection during intraoperative cardioplegic arrest [174].

The risk factors for POBAs may be classified as preoperative, operative, and postoperative factors. Preoperative risk factors include age >75 years, the use of rate lowering cardiac medications (e.g., beta-blockers, calcium channel blockers, digoxin, and antiarrhythmic drugs), the presence of conduction system disease preoperatively, right bundle branch block (RBBB) or left bundle branch block (LBBB), first-degree AV block or left anterior fascicular block (LAFB) [169171, 175178].

Operative risk factors include myocardial ischemia, inadequate cardiac protection during surgery, and direct surgical injury to conduction system, prolonged CPB time and cross-clamp time, and reoperation [171, 172, 174, 179, 180].

Postoperative risk factors include postoperative conduction disturbances and high-grade AV block [174, 175, 181].

5.3. Clinical course and management

Temporary electrical pacing may be required in symptomatic bradycardias. It is common practice nowadays to place temporary epicardial atrial and ventricular pacing wires placed at the time of surgery to facilitate temporary pacing when needed. In some cases, as mentioned above, the conduction defect does not revert, and permanent pacing may be necessary. Chronotropic medications, such as theophylline or aminophylline, have been used for sinus bradycardia after transplantation to improve SSS [182] or high-grade AVB [183] and may decrease the need for permanent pacing but its long-term effect is not encouraging.

The challenge with POBAs is often to determine when to implant the PPM as the sinus node function or AV conduction may recover in some patients. Recovery of conduction system is common with long-term follow-up. Only 30–40% of patients with a permanent pacemaker due to sinus node disease remain pacemaker dependent. However, the rate of recovery is less in patients with postoperative AVB, as 65–100% of patients with complete heart block, remain pacemaker dependent. Currently, the usual practice is to implant a PPM if postoperative symptomatic complete AVB or severe sinus node dysfunction persists longer than 5–7 days [184]. PPM implantation may be considered earlier if the underlying intrinsic rhythm is absent or temporary pacing leads fail.

Advertisement

6. Conclusion

Arrhythmias are common after CABG. Although tachyarrhythmias are frequent, they are usually transient and have a benign course. POAF represents the most frequently observed ATs. VAs are less common but have an adverse impact on the short and long-term outcome. POTAs management includes optimization of the patient’s condition, controlling patient’s risk factors, and careful attention to hemodynamic changes during surgery with using appropriate anesthesia. Postoperatively, it is important to correct reversible arrhythmia predisposing factors, followed by specific therapy based on the arrhythmia type and its hemodynamic effect.

On the other hand, bradyarrhythmias are also frequently observed after cardiac surgery. However, most of the conduction disturbances are transient and recovered spontaneously. PPM implantation may be required in patients with persistent symptomatic bradycardia due to sick sinus syndrome or second degree type 2, third degree, or high-grade AV block.

References

  1. 1. Herzog L, Lynch C. Arrhythmias accompanying cardiac surgery. In: Lynch C, editor. Clinical Cardiac Electrophysiology. 3rd ed. Philadelphia, PA, USA: JB Lippincott; 1994. pp. 231-258.
  2. 2. Creswell LL, Schuessler RB, Rosenbloom M, Cox JL. Hazards of postoperative atrial arrhythmias. The Annals of Thoracic Surgery. 1993;56:539-549. DOI: https://doi.org/10.1016/0003-4975(93)90894-N
  3. 3. Mathew JP, Fontes ML, Tudor IC, Ramsay J, Duke P, Mazer CD, Barash PG, Hsu PH, Mangano DT, Investigators of the Ischemia Research and Education Foundation, Multicenter Study of Perioperative Ischemia Research Group. A multicenter risk index for atrial fibrillation after cardiac surgery. JAMA. 2004;291:1720-1729. DOI: 10.1001/jama.291.14.1720
  4. 4. Steinbach K, Merl O, Frohner K, Hief C, Nurnberg M, Kalten-brunner W, Podczeck A, Wessely E. Hemodynamics during ventricular tachyarrhythmias. American Heart Journal. 1994;127:1102-1106. DOI: http://dx.doi.org/10.1016/0002-8703(94)90095-7
  5. 5. Fenelon G, Wigjns W, Andries E, Brugada P. Tachycardiomyopathy: Mechanisms and clinical implications. PACE. 1996;19:95-106. DOI: 10.1111/j.1540-8159.1996.tb04796.x
  6. 6. Baig MW, Perrins EJ. The hemodynamics of cardiac pacing. Progress in Cardiovascular Diseases. 1991;33:283-298. DOI: https://doi.org/10.1016/0033-0620(91)90021-D
  7. 7. Peretto G, Durante A, Limite LR, Cianflone D. Postoperative arrhythmias after cardiac surgery: Incidence, risk factors, and therapeutic management. Cardiology Research and Practice. 2014;2014:615987. DOI: 10.1155/2014/615987
  8. 8. Diodato M, Chedrawy EG. Coronary artery bypass graft surgery: The past, present, and future of myocardial revascularisation. Surgery Research and Practice. 2014;2014:726158.DOI: 10.1155/2014/726158
  9. 9. Møller CH, Penninga L, Wetterslev J, Steinbruchel DA, Gluud C. Off-pump versus on-pump coronary artery bypass grafting for ischaemic heart disease. Cochrane Database of Systematic Reviews. 2012 March 14;3:CD007224. DOI: 10.1002/14651858.CD007224.pub2
  10. 10. Mack M, Acuff T, Yong P, Jett GK, Carter D. Minimally invasive thoracoscopically assisted coronary artery bypass surgery. European Journal of Cardio-Thoracic Surgery. 1997;12:20-24. DOI: https://doi.org/10.1016/S1010-7940(97)00141-3
  11. 11. Enríquez F, Jiménez A. Post-operative tachyarrhythmias in adult cardiac surgery: Prophylaxis. Cirugia Cardiovascular. 2010;17:259-274. DOI: Https://doi.org/10.1016/S1134-0096(10)70100-5 (in Spanish: Taquiarritmias postoperatorias en la cirugía cardíaca del adulto. Profilaxis)
  12. 12. Zaman AG, Archbold RA, Helft G, Atrial Paul EA, Curzen NP, Mills PG. Atrial fibrillation after coronary artery bypass surgery: A model for preoperative risk stratification. Circulation. 2000;101:1403-1408. DOI: https://doi.org/10.1161/01.CIR.101.12.1403
  13. 13. Leitch JW, Thomson D, Baird DK, Harris PJ. The importance of age as a predictor of atrial fibrillation and flutter after coronary artery bypass grafting. The Journal of Thoracic and Cardiovascular Surgery. 1990;100:338-342 PMID: 2391970
  14. 14. Premaratne S, Premaratne ID, Fernando ND, Williams L, Hasaniya NW. Atrial fibrillation and flutter following coronary artery bypass graft surgery: A retrospective study and review. JRSM Cardiovascular Disease. 2016;5:2048004016634149. DOI: 10.1177/2048004016634149
  15. 15. Mendes LA, Connelly GP, McKenney PA, Podrid PJ, Cupples LA, Shemin RJ, Ryan TJ, Davidoff R. Right coronary artery stenosis: An independent predictor of atrial fibrillation after coronary artery bypass surgery. Journal of the American College of Cardiology. 1995;25:198-202. DOI: https://doi.org/10.1016/0735-1097(94)00329-O
  16. 16. Helgadottir S, Sigurdsson MI, Ingvarsdottir IL, Arnar DO, Gudbjartsson T. Atrial fibrillation following cardiac surgery: Risk analysis and long-term survival. Journal of Cardiothoracic Surgery. 2012;7:87. DOI: 10.1186/1749-8090-7-87
  17. 17. Kolvekar S, D’Souza A, Akhtar P, Reek C, Garratt C, Spyt T. Role of atrial ischaemia in development of atrial fibrillation following coronary artery bypass surgery. European Journal of Cardio-Thoracic Surgery. 1997;11:70-75. DOI: 10.1016/S1010-7940(96)01095-0
  18. 18. Wazni OM, Martin DO, Marrouche NF, Latif AA, Ziada K, Shaaraoui M, Almahameed S, Schweikert RA, Saliba WI, Gillinov AM, Tang WH, Mills RM, Francis GS, Young JB, Natale A. Plasma B-type natriuretic peptide levels predict postoperative atrial fibrillation in patients undergoing cardiac surgery. Circulation. 2004;110:124-127. DOI: https://doi.org/10.1161/01.CIR.0000134481.24511.BC
  19. 19. Zacharias A, Schwann TA, Riordan CJ, Durham SJ, Shah AS, Habib RH. Obesity and risk of new-onset atrial fibrillation after cardiac surgery. Circulation. 2005;112:3247-3255 https://doi.org/10.1161/CIRCULATIONAHA.105.553743
  20. 20. Maesen B, Nijs J, Maessen J, Allessie M, Schotten U. Post-operative atrial fibrillation: A maze of mechanisms. Europace. 2012;14:159-174. DOI: https://doi.org/10.1093/europace/eur208
  21. 21. Warltier DC, Laffey JG, Boylan JF, Cheng DCH. The systemic inflammatory response to cardiac surgery: Implications for the anesthesiologist. Anesthesiology. 2002;97:215-252 PMID: 12131125
  22. 22. Bulow NMH, Colpo E, Duarte MF, Correa EFM, Schlosser RS, Lauda A, Kade IJ, Rocha JBT. Inflammatory response in patients under coronary artery bypass grafting surgery and clinical implications: A review of the relevance of Dexmedetomidine use. ISRN Anesthesiology. 2014;2014:1-28 http://dx.doi.org/10.1155/2014/905238
  23. 23. Prondzinsky R, Knüpfer A, Loppnow H, Redling F, Lehmann DW, Stabenow I, Witthaut  R, Unverzagt S, Radke J, Zerkowski HR, Werdan K. Surgical trauma affects the proinflammatory status after cardiac surgery to a higher degree than cardiopulmonary bypass. The Journal of Thoracic and Cardiovascular Surgery. 2005;129:760-766. DOI: https://doi.org/10.1016/j.jtcvs.2004.07.052
  24. 24. Angelini GD, Penny WJ, el-Ghamary F, West RR, Butchart EG. The incidence and significance of early pericardial effusion after open heart surgery. European Journal of Cardio-Thoracic Surgery. 1987;1:165-168. DOI: https://doi.org/10.1016/1010-7940(87)90034-0
  25. 25. Stamou SC, Dangas G, Hill PC, Pfister AJ, Dullum MK, Boyce SW, Bafi AS, Garcia JM, Corso PJ. Atrial fibrillation after beating heart surgery. The American Journal of Cardiology. 2000;86:64-67. DOI: https://doi.org/10.1016/S0002-9149(00)00829-8
  26. 26. Haïssaguerre M, Jaïs P, Shah DC, Takahashi A, Hocini M, Quiniou G, Garrigue S, Le Mouroux A, Le Métayer P, Clémenty J. Spontaneous initiation of atrial fibrillation by ectopic beats originating in the pulmonary veins. The New England Journal of Medicine. 1998;339:659-666. DOI: 10.1056/NEJM199809033391003
  27. 27. Vinten-Johansen J. Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovascular Research. 2004;61:481-497. DOI: 10.1016/j.cardiores.2003.10.011
  28. 28. Butler J, Parker D, Pillai R, Westaby S, Shale DJ, Rocker GM. Effects of cardiopulmonary bypass on systemic release of neutrophil elastase and tumour necrosis factor. The Journal of Thoracic and Cardiovascular Surgery. 1993;105:25-30 PMID:8419705
  29. 29. Wakayama F, Fukuda I, Suzuki Y, Kondo N. Neutrophil elastase inhibitor, sivelestat, attenuates acute lung injury after cardiopulmonary bypass in the rabbit endotoxemia model. The Annals of Thoracic Surgery. 2007;83:153-160. DOI: 10.1016/j.athoracsur.2006.08.023
  30. 30. Atlee JL. Perioperative cardiac dysrhythmias: Diagnosis and management. Anesthesiology. 1997;86:1397-1424
  31. 31. Salazar C, Frishman W, Friedman S, Patel J, Lin YT, Oka Y, Frater RW, Becker RM. 𝛽-blockade therapy for supraventricular tachyarrhythmias after coronary surgery: A propranolol withdrawal syndrome? Angiology. 1979;30:816-819. DOI: 10.1177/000331977903001204
  32. 32. Deliargyris EN, Raymond RJ, Guzzo JA, Dehmer GJ, Smith SC, Weiner MS, Roberts CS. Preoperative factors predisposing to early postoperative atrial fibrillation after isolated coronary artery bypass grafting. The American Journal of Cardiology. 2000;85:763-764. DOI: http://dx.doi.org/10.1016/S0002-9149(99)00857-7
  33. 33. Rubin DA, Nieminski KE, Reed GE, Herman MV. Predictors, prevention, and long-term prognosis of atrial fibrillation after coronary artery bypass graft operations. The Journal of Thoracic and Cardiovascular Surgery. 1987;94:331-335 PMID: 3306163
  34. 34. Tisdale JE, Patel R, Webb CR, Borzak S, Zarowitz BJ. Electrophysiologic and proarrhythmic effects of intravenous inotropic agents. Progress in Cardiovascular Diseases. 1995;38:167-180. DOI: http://dx.doi.org/10.1016/S0033-0620(05)80005-2
  35. 35. Williams JB, Hernandez AF, Li S, Dokholyan RS, O'Brien SM, Smith PK, Ferguson TB, Peterson ED. Postoperative inotrope and vasopressor use following CABG: Outcome data from the CAPS-care study. Journal of Cardiac Surgery. 2011;26:572-578. DOI: 10.1111/j.1540-8191.2011.01301.x
  36. 36. Haghjoo M. Pharmacological and nonpharmacological prevention of atrial fibrillation after coronary artery bypass surgery. The Journal of Tehran Heart Center. 2012;7:2-9 PMCID: PMC3466882
  37. 37. Wahr JA, Parks R, Boisvert D, Comunale M, Fabian J, Ramsay J, Mangano DT. Preoperative serum potassium levels and perioperative outcomes in cardiac surgery patients. Multicenter study of perioperative ischemia research group. JAMA. 1999;281:2203-2210. DOI: 10.1001/jama.281.23.2203
  38. 38. Luo X, Pan Z, Shan H, Xiao J, Sun X, Wang N, Lin H, Xiao L, Maguy A, Qi XY, Li Y, Gao X, Dong D, Zhang Y, Bai Y, Ai J, Sun L, Lu H, Luo XY, Wang Z, Lu Y, Yang B, Nattel S. MicroRNA-26 governs profibrillatory inward-rectifier potassium current changes in atrial fibrillation. The Journal of Clinical Investigation. 2013;123:1939-1951. DOI: 10.1172/JCI62185
  39. 39. Pinski SL. Potassium replacement after cardiac surgery: It is not time to change practice, yet. Critical Care Medicine. 1999;27:2581-2582 PMID: 10579291
  40. 40. England MR, Gordon G, Salem M, Chernow B. Magnesium administration and dysrhythmias after cardiac surgery: A placebo-controlled, double-blind, randomized trial. JAMA. 1992;268:2395-2402. DOI: 10.1001/jama.1992.03490170067027
  41. 41. Aglio LS, Stanford GC, Maddi R, Boyd III JL, Nussbaum S, Chernow B. Hypomagnesemia is common following cardiac surgery. Journal of Cardiothoracic and Vascular Anesthesia. 1991;5:201-208. DOI: http://dx.doi.org/10.1016/1053-0770(91)90274-W
  42. 42. Singh S, Johnson PI, Lee RE, Orfei E, Lonchyna VA, Sullivan HJ, Montoya A, Tran H, Wehrmacher WH, Wurster RD. Topography of cardiac ganglia in the adult human heart. The Journal of Thoracic and Cardiovascular Surgery. 1996;112:943-953. DOI: 10.1016/S0022-5223(96)70094-6
  43. 43. Quan KJ, Lee JH, Geha AS, Biblo LA, Van Hare GF, Mackall JA, Carlson MD. Characterization of sinoatrial parasympathetic innervation in humans. Journal of Cardiovascular Electrophysiology. 1999;10:1060-1065. DOI: 10.1111/j.1540-8167.1999.tb00278.x
  44. 44. Quan KJ, Lee JH, Van Hare GF, Biblo LA, Mackall JA, Carlson MD. Identification and characterization of atrioventricular parasympathetic innervation in humans. Journal of Cardiovascular Electrophysiology. 2002;13:735-739. DOI: 10.1046/j.1540-8167.2002.00735.x
  45. 45. Carlson MD, Geha AS, Hsu J, Martin PJ, Levy MN, Jacobs G, Waldo AL. Selective stimulation of parasympathetic nerve fibers to the human sinoatrial node. Circulation. 1992;85:1311-1317. DOI: https://doi.org/10.1161/01.CIR.85.4.1311
  46. 46. Cummings JE, Gill I, Akhrass R, Dery Biblo LA, Quan KJ. Preservation of the anterior fat pad paradoxically decreases the incidence of postoperative atrial fibrillation in humans. Journal of the American College of Cardiology. 2004;17(43):994-1000. DOI: 10.1016/j.jacc.2003.07.055
  47. 47. CM W, Sander S, Coleman CI, Gallagher R, Takata H, Humphrey C, Henyan N, Gillespie EL, Kluger J. Impact of epicardial anterior fat pad retention on postcardiothoracic surgery atrial fibrillation incidence: The AFIST-III study. Journal of the American College of Cardiology. 2007;49:298-303. DOI: 10.1016/j.jacc.2006.10.033
  48. 48. Mitchell LB. Incidence, timing and outcome of atrial tachyarrhythmias after cardiac surgery. In: Steinberg JS, editor. Atrial Fibrillation after Cardiac Surgery. Boston: Kluwer Academic Publishers; 2000. pp. 37-50.
  49. 49. Maisel WH, Rawn JD, Stevenson WG. Atrial fibrillation after cardiac surgery. Annals of Internal Medicine. 2001;135:1061-1073. DOI: 10.7326/0003-4819-135-12-200112180-00010
  50. 50. Blomstrom Lundqvist C. Post CABG atrial fibrillation: What are the incidence, predictors, treatment, and long-term outcome? In: Raviele A, editor. Cardiac Arrhythmias 2005. Venice, Italy: Springer; 2005. pp. 131-136.
  51. 51. Pavri BB, O'Nunain SS, Newell JB, Ruskin JN, William G. Prevalence and prognostic significance of atrial arrhythmias after orthotopic cardiac transplantation. Journal of the American College of Cardiology. 1995;25:1673-1680. DOI: https://doi.org/10.1016/0735-1097(95)00047-8
  52. 52. Mathew JP, Parks R, Savino JS, Friedman AS, Koch C, Mangano DT, Browner WS. Atrial fibrillation following coronary artery bypass graft surgery: Predictors, outcomes, and resource utilization. JAMA. 1996;276:300-306. DOI: 10.1001/jama.1996.03540040044031
  53. 53. Echahidi N, Pibarot P, O’Hara G, Mathieu P. Mechanisms, prevention, and treatment of atrial fibrillation after cardiac surgery. Journal of the American College of Cardiology. 2008;51:793-801. DOI: 10.1016/j.jacc. 2007.10.043
  54. 54. Shingu Y, Kubota S, Wakasa S, Ooka T, Tachibana T, Matsui Y. Postoperative atrial fibrillation: Mechanism, prevention, and future perspective. Surgery Today. 2012;42:819-824. DOI: 10.1007/s00595-012-0199-4
  55. 55. Aranki SF, Shaw DP, Adams DH, Rizzo RJ, Couper GS, VanderVliet M, Collins JJ Jr, Cohn LH, Burstin HR. Current trends and impact on hospital resources. Circulation. 1996;94:390-397. DOI: https://doi.org/10.1161/01.CIR.94.3.390
  56. 56. Soucier RJ, Mirza S, Abordo MG, Berns E, Dalamagas HC, Hanna A, Silverman DI. Predictors of conversion of atrial fibrillation after cardiac operation in the absence of class I or III antiarrhythmic medications. The Annals of Thoracic Surgery. 2001;72:694-697. DOI: http://dx.doi.org/10.1016/S0003-4975(01)02817-X
  57. 57. Lee JK, Klein GJ, Krahn AD, Yee R, Zarnke K, Simpson C, Skanes A, Spindler B. Rate-control versus conversion strategy in postoperative atrial fibrillation: A prospective, randomized pilot study. American Heart Journal. 2000;140:871-877. DOI: 10.1067/mhj.2000.111104
  58. 58. Landymore RW, Howell F. Recurrent atrial arrhythmias following treatment for postoperative atrial fibrillation after coronary bypass operations. European Journal of Cardio-Thoracic Surgery. 1991;5:436-439. DOI: https://doi.org/10.1016/1010-7940(91)90191-L
  59. 59. Kowey PR, Stebbins D, Igidbashian L, Goldman SM, Sutter FP, Rials SJ, Marinchak RA. Clinical outcome of patients who develop PAF after CABG surgery. Pacing and Clinical Electrophysiology. 2001;24:191-193. DOI: 10.1046/j.1460-9592.2001.00191.x
  60. 60. Villareal RP, Hariharan R, Liu BC, Kar B, Lee VV, Elayda M, Lopez JA, Rasekh A, Wilson JM, Massumi A. Postoperative atrial fibrillation and mortality after coronary artery bypass surgery. Journal of the American College of Cardiology. 2004;43:742-748. DOI: 10.1016/j.jacc.2003.11.023
  61. 61. Fuller JA, Adams GG, Buxton B. Atrial fibrillation after coronary artery bypass grafting: Is it a disorder of the elderly? The Journal of Thoracic and Cardiovascular Surgery. 1989;97:821-825 PMID: 2566713
  62. 62. Lahtinen J, Biancari F, Salmela E, Mosorin M, Satta J, Rainio P, Rimpiläinen J, Lepojärvi M, Juvonen T. Postoperative atrial fibrillation is a major cause of stroke after on-pump coronary artery bypass surgery. The Annals of Thoracic Surgery. 2004;77:1241-1244. DOI: 10.1016/j.athoracsur.2003.09.077
  63. 63. Reed GL, III, Singer DE, Picard EH, DeSanctis RW. Stroke following coronary-artery bypass surgery. A case-control estimate of the risk from carotid bruits. The New England Journal of Medicine. 1988;319:1246-1250. DOI: 10.1056/NEJM198811103191903
  64. 64. Newman MF, Wolman R, Kanchuger M, Marschall K, Mora-Mangano C, Roach G, Smith LR, Aggarwal A, Nussmeier N, Herskowitz A, Mangano DT. Multicenter preoperative stroke risk index for patients undergoing coronary artery bypass graft surgery. Multicenter study of perioperative ischemia (McSPI) research group. Circulation. 1996;94(9 Suppl):II74-II80 PMID: 8901723
  65. 65. Mickleborough LL, Walker PM, Takagi Y, Ohashi M, Ivanov J, Tamariz M. Risk factors for stroke in patients undergoing coronary artery bypass grafting. The Journal of Thoracic and Cardiovascular Surgery. 1996;112:1250-1259. DOI: 10.1016/S0022-5223(96)70138-1
  66. 66. Hogue CW Jr, Murphy SF, Schechtman KB, Dávila-Román VG. Risk factors for early or delayed stroke after cardiac surgery. Circulation. 1999;100:642-647. DOI: https://doi.org/10.1161/01.CIR.100.6.642
  67. 67. Kim MH, Deeb GM, Morady F, Bruckman D, Hallock LR, Smith KA, Karavite DJ, Bolling SF, Pagani FD, Wahr JA, Sonnad SS, Kazanjian PE, Watts C, Williams M, Eagle KA. Effect of postoperative atrial fibrillation on length of stay after cardiac surgery (the postoperative atrial fibrillation in cardiac surgery study [PACS2]). The American Journal of Cardiology. 2001;87:881-885. DOI: http://dx.doi.org/10.1016/S0002-9149(00)01530-7
  68. 68. Lauer MS, Eagle KA, Buckley MJ, DeSanctis RW. Atrial fibrillation following coronary artery bypass surgery. Progress in Cardiovascular Diseases. 1989;31:367-378. DOI: http://dx.doi.org/10.1016/0033-0620(89)90031-5
  69. 69. Wyse DG, Waldo AL, DiMarco JP, Domanski MJ, Rosenberg Y, Schron EB, Kellen JC, Greene HL, Mickel MC, Dalquist JE. Atrial fibrillation follow-up investigation of rhythm management (AFFIRM) investigators a comparison of rate control and rhythm control in patients with atrial fibrillation. The New England Journal of Medicine. 2002;347:1825-1833. DOI: 10.1056/NEJMoa021328.
  70. 70. Jongnarangsin K, Oral H. Postoperative atrial fibrillation. Cardiology Clinics. 2009;27:69-78. DOI: 10.1016/j.ccl.2008.09.011
  71. 71. Mayson SE, Greenspon AJ, Adams S, Decaro MV, Sheth M, Weitz HH, Whellan DJ. The changing face of postoperative atrial fibrillation prevention: A review of current medical therapy. Cardiology in Review. 2007;15:231-241. DOI: 10.1097/CRD.0b013e31813e62bb
  72. 72. Al-Khatib SM, Hafley G, Harrington RA, Mack MJ, Ferguson TB, Peterson ED, Califf RM, Kouchoukos NT, Alexander JH. Patterns of management of atrial fibrillation complicating coronary artery bypass grafting: Results from the PRoject of ex-vivo vein graft ENgineering via transfection IV (PREVENT-IV) trial. American Heart Journal. 2009;158:792-798. DOI: 10.1016/j.ahj.2009.09.003
  73. 73. Gillinov AM, Bagiella E, Moskowitz AJ, Raiten JM, Groh MA, Bowdish ME, Ailawadi G, Kirkwood KA, Perrault LP, Parides MK, Smith RL II, Kern JA, Dussault G, Hackmann AE, Jeffries NO, Miller MA, Taddei-Peters WC, Rose EA, Weisel RD, Williams DL, Mangusan RF, Argenziano M, Moquete EG, O'Sullivan KL, Pellerin M, Shah KJ, Gammie JS, Mayer ML, Voisine P, Gelijns AC, O'Gara PT, Mack MJ, CTSN. Rate control versus rhythm control for atrial fibrillation after cardiac surgery. The New England Journal of Medicine. 2016;374:1911-1921. DOI: 10.1056/NEJMoa1602002.
  74. 74. January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC Jr, Conti JB, Ellinor PT, Ezekowitz MD, Field ME, Murray KT, Sacco RL, Stevenson WG, Tchou PJ, Tracy CM, Yancy CW; ACC/AHA Task Force Members. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: Executive summary: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. Circulation 2014;130:2071-2104. DOI: 10.1161/CIR.0000000000000040
  75. 75. Kirchhof P, Benussi S, Kotecha D, Ahlsson A, Atar D, Casadei B, Castellá M, Diener HC, Heidbuchel H, Hendriks J, Hindricks G, Manolis AS, Oldgren J, Alexandru Popescu B, Schotten U, Van Putte B, Vardas P. 2016 ESC guidelines for the management of atrial fibrillation developed in collaboration with EACTS. European Heart Journal. 2016 Oct 7;37(38):2893-2962. DOI: 10.1093/eurheartj/ehw210
  76. 76. Clemo HF, Wood MA, Gilligan DM, Ellenbogen KA. Intravenous amiodarone for acute heart rate control in the critically ill patient with atrial tachyarrhythmias. The American Journal of Cardiology. 1998;81:594-598. DOI: http://dx.doi.org/10.1016/S0002-9149(97)00962-4
  77. 77. Daoud EG, Strickberger SA, Man KC, Goyal R, Deeb GM, Bolling SF, Pagani FD, Bitar C, Meissner MD, Morady F. Preoperative amiodarone as prophylaxis against atrial fibrillation after heart surgery. The New England Journal of Medicine. 1997;337:1785-1791. DOI: 10.1056/NEJM199712183372501
  78. 78. Dunning J, Treasure T, Versteegh M, Nashef SA, EACTS Audit and Guidelines Committee. Guidelines on the prevention and management of de novo atrial fibrillation after cardiac and thoracic surgery. European Journal of Cardiothoracic Surgery. 2006;30:852-872. DOI: 10.1016/j.ejcts.2006.09.003
  79. 79. Heldal M, Atar D. Pharmacological conversion of recent-onset atrial fibrillation: A systematic review. Scandinavian Cardiovascular Journal. Supplement. 2013;47:2-10. DOI: 10.3109/14017431.2012.740572
  80. 80. Kowey PR, Dorian P, Mitchell LB, Pratt CM, Roy D, Schwartz PJ, Sadowski J, Sobczyk D, Bochenek A, Toft E, Atrial Arrhythmia Conversion Trial Investigators. Vernakalant hydrochloride for the rapid conversion of atrial fibrillation after cardiac surgery: A randomized, double-blind, placebo-controlled trial. Circulation. Arrhythmia and Electrophysiology. 2009;2:652-659. DOI: 10.1161/CIRCEP.109.870204
  81. 81. VanderLugt JT, Mattioni T, Denker S, Torchiana D, Ahern T, Wakefield LK, Perry KT, Kowey PR. Efficacy and safety of ibutilide fumarate for the conversion of atrial arrhythmias after cardiac surgery. Circulation. 1999;100:369-375. DOI: https://doi.org/10.1161/01.CIR.100.4.369
  82. 82. Gomes JA, Ip J, Santoni-Rugiu F, Mehta D, Ergin A, Lansman S, Pe E, Newhouse TT, Chao S. Oral d,l sotalol reduces the incidence of post-operative atrial fibrillation in coronary artery bypass surgery patients: A randomized, double-blind, placebo-controlled study. Journal of the American College of Cardiology. 1999;34:334-339. DOI: https://doi.org/10.1016/S0735-1097(99)00213-2
  83. 83. El-Chami MF, Kilgo P, Thourani V, Lattouf OM, Delurgio DB, Guyton RA, Leon AR, Puskas JD. New-onset atrial fibrillation predicts long-term mortality after coronary artery bypass graft. Journal of the American College of Cardiology. 2010;55:1370-1376. DOI: 10.1016/j.jacc.2009.10.058
  84. 84. Anderson E, Dyke C, Levy JH. Anticoagulation strategies for the management of postoperative atrial fibrillation. Clinics in Laboratory Medicine. 2014;34:537-561. DOI: 10.1016/j.cll.2014.06.012
  85. 85. Arsenault KA, Yusuf AM, Crystal E, Healey JS, Morillo CA, Nair GM, Whitlock RP. Interventions for preventing post-operative atrial fibrillation in patients undergoing heart surgery. Cochrane Database of Systematic Reviews. 2013;1:CD003611. DOI: 10.1002/14651858.CD003611.pub3
  86. 86. Khan MF, Wendel CS, Movahed MR. Prevention of post-coronary artery bypass grafting (CABG) atrial fibrillation: Efficacy of prophylactic beta-blockers in the modern era: A meta-analysis of latest randomized controlled trials. Annals of Noninvasive Electrocardiology. 2013;18:58-68. DOI: 10.1111/anec.12004
  87. 87. Burgess DC, Kilborn MJ, Keech AC. Interventions for prevention of post-operative atrial fibrillation and its complications after cardiac surgery: A meta-analysis. European Heart Journal. 2006;27:2846-2857. DOI: 10.1093/eurheartj/ehl272
  88. 88. Chatterjee S, Sardar P, Mukherjee D, Lichstein E, Aikat S. Timing and route of amiodarone for prevention of postoperative atrial fibrillation after cardiac surgery: A network regression meta-analysis. Pacing and Clinical Electrophysiology. 2013;36:1017-1023. DOI: 10.1111/pace.12140
  89. 89. Zhu J, Wang C, Gao D, Zhang C, Zhang Y, Lu Y, Gao Y. Meta-analysis of amiodarone versus beta-blocker as a prophylactic therapy against atrial fibrillation following cardiac surgery. Internal Medicine Journal. 2012;42:1078-1087. DOI: 10.1111/j.1445-5994.2012.02844.x
  90. 90. Koniari I, Apostolakis E, Rogkakou C, Baikoussis NG, Dougenis D. Pharmacologic prophylaxis for atrial fibrillation following cardiac surgery: A systematic review. Journal of Cardiothoracic Surgery. 2010;5:121. DOI: 10.1186/1749-8090-5-121
  91. 91. Shepherd J, Jones J, Frampton GK, Tanajewski L, Turner D, Price A. Intravenous magnesium sulphate and sotalol for prevention of atrial fibrillation after coronary artery bypass surgery: A systematic review and economic evaluation. Health Technology Assessment. 2008;12:iii-iv, ix-95(1-49). DOI: https://dx.doi.org/10.3310/hta12280
  92. 92. Imazio M, Brucato A, Ferrazzi P, Rovere ME, Gandino A, Cemin R, Ferrua S, Belli R, Maestroni S, Simon C, Zingarelli E, Barosi A, Sansone F, Patrini D, Vitali E, Trinchero R, Spodick DH, Adler Y, COPPS Investigators. Colchicine reduces postoperative atrial fibrillation: Results of the colchicine for the prevention of the Postpericardiotomy syndrome (COPPS) atrial fibrillation substudy. Circulation. 2011;124:2290-2295. DOI: 10.1161/CIRCULATIONAHA.111.026153
  93. 93. Zheng Z, Jayaram R, Jiang L, Emberson J, Zhao Y, Li Q, Du J, Guarguagli S, Hill M, Chen Z, Collins R, Casadei B. Perioperative Rosuvastatin in cardiac surgery. The New England Journal of Medicine. 2016;374:1744-1753. DOI: 10.1056/NEJMoa1507750
  94. 94. Liakopoulos OJ, Kuhn EW, Slottosch I, Wassmer G, Wahlers T. Preoperative statin therapy for patients undergoing cardiac surgery. Cochrane Database of Systematic Reviews. 2012;4:Cd008493. DOI: 10.1002/14651858.CD008493.pub2
  95. 95. Fauchier L, Clementy N, Babuty D. Statin therapy and atrial fibrillation: Systematic review and updated meta-analysis of published randomized controlled trials. Current Opinion in Cardiology. 2013;28:7-18. DOI: 10.1097/HCO.0b013e32835b0956
  96. 96. Zheng H, Xue S, ZL H, Shan JG, Yang WG. The use of statins to prevent postoperative atrial fibrillation after coronary artery bypass grafting: A meta-analysis of 12 studies. Journal of Cardiovascular Pharmacology. 2014 Sep;64(3):285-292. DOI: 10.1097/FJC.0000000000000102
  97. 97. Mathew JP, Fontes ML, Tudor IC, Ramsay J, Duke P, Mazer CD, Barash PG, Hsu PH, Mangano DT, Investigators of the Ischemia Research and Education Foundation, Multicenter Study of Perioperative Ischemia Research Group. A multicenter risk index for atrial fibrillation after cardiac surgery. Journal of the American Medical Association. 2004;14(291):1720-1729. DOI: 10.1001/jama.291.14.1720
  98. 98. Cook RC, Yamashita MH, Kearns M, Ramanathan K, Gin K, Humphries KH. Prophylactic magnesium does not prevent atrial fibrillation after cardiac surgery: A meta-analysis. The Annals of Thoracic Surgery. 2013;95:533-541. DOI: 10.1016/j.athoracsur.2012.09.008
  99. 99. De Oliveira GS, Jr Knautz JS, Sherwani S, McCarthy RJ. Systemic magnesium to reduce postoperative arrhythmias after coronary artery bypass graft surgery: A meta-analysis of randomized controlled trials. Journal of Cardiothoracic and Vascular Anesthesia. 2012;26:643-650. DOI: 10.1053/j.jvca.2012.03.012
  100. 100. Costanzo S, di Niro V, Di Castelnuovo A, Gianfagna F, Donati MB, de Gaetano G, Iacoviello L. Prevention of postoperative atrial fibrillation in open heart surgery patients by preoperative supplementation of n-3 polyunsaturated fatty acids: An updated meta-analysis. The Journal of Thoracic and Cardiovascular Surgery. 2013;146:906-911. DOI: 10.1016/j.jtcvs.2013.03.015
  101. 101. Farquharson AL, Metcalf RG, Sanders P, Stuklis R, Edwards JR, Gibson RA, Cleland LG, Sullivan TR, James MJ, Young GD. Effect of dietary fish oil on atrial fibrillation after cardiac surgery. The American Journal of Cardiology. 2011;108:851-856. DOI: 10.1016/j.amjcard.2011.04.036
  102. 102. Heidarsdottir R, Arnar DO, Skuladottir GV, Torfason B, Edvardsson V, Gottskalksson G, Palsson R, Indridason OS. Does treatment with n-3 polyunsaturated fatty acids prevent atrial fibrillation after open heart surgery? Europace. 2010;12:356-363. DOI: 10.1093/europace/eup429
  103. 103. Mariani J, Doval HC, Nul D, Varini S, Grancelli H, Ferrante D, Tognoni G, Macchia A. N-3 polyunsaturated fatty acids to prevent atrial fibrillation: Updated systematic review and meta-analysis of randomized controlled trials. Journal of the American Heart Association. 2013;2:e005033. DOI: 10.1161/JAHA.112.005033
  104. 104. Rodrigo R, Korantzopoulos P, Cereceda M, Asenjo R, Zamorano J, Villalabeitia E, Baeza C, Aguayo R, Castillo R, Carrasco R, Gormaz JG. A randomized controled trial to prevent post-operative atrial fibrillation by antioxidant reinforcement. Journal of the American College of Cardiology. 2013;62:1457-1465. DOI: 10.1016/j.jacc.2013.07.014
  105. 105. Saravanan P, Bridgewater B, West AL, O’Neill SC, Calder PC, Davidson NC. Omega-3 fatty acid supplementation does not reduce risk of atrial fibrillation after coronary artery bypass surgery: A randomized, double-blind, placebo-controlled clinical trial. Circulation. Arrhythmia and Electrophysiology. 2010;3:46-53. DOI: 10.1161/CIRCEP.109.899633
  106. 106. Wu JH, Marchioli R, Silletta MG, Macchia A, Song X, Siscovick DS, Harris WS, Masson S, Latini R, Albert C, Brown NJ, Lamarra M, Favaloro RR, Mozaffarian D. Plasma phospholipid omega-3 fatty acids and incidence of postoperative atrial fibrillation in the OPERA trial. Journal of the American Heart Association. 2013;2:e000397. DOI: 10.1161/JAHA.113.000397
  107. 107. Xin W, Wei W, Lin Z, Zhang X, Yang H, Zhang T, Li B, Mi S. Fish oil and atrial fibrillation after cardiac surgery: A meta-analysis of randomized controlled trials. PloS One. 2013;8:e72913. DOI: 0.1371/journal.pone.0072913
  108. 108. Zhang B, Zhen Y, Tao A, Bao Z, Zhang G. Polyunsaturated fatty acids for the prevention of atrial fibrillation after cardiac surgery: An updated meta-analysis of randomized controlled trials. Journal of Cardiology. 2014;63:53-59. DOI: 10.1016/j.jjcc.2013.06.014
  109. 109. Ho KM, Tan JA. Benefits and risks of corticosteroid prophylaxis in adult cardiac surgery a dose-response meta-analysis. Circulation. 2009;119:1853-1866. DOI: 10.1161/CIRCULATIONAHA.108.848218
  110. 110. Cappabianca G, Rotunno C, de Luca Tupputi Schinosa L, Ranieri VM, Paparella D. Protective effects of steroids in cardiac surgery: A meta-analysis of randomized double-blind trials. Journal of Cardiothoracic and Vascular Anesthesia. 2011;25:156-165. DOI: 10.1053/j.jvca.2010.03.015
  111. 111. Viviano A, Kanagasabay R, Zakkar M. Is perioperative corticosteroid administration associated with a reduced incidence of postoperative atrial fibrillation in adult cardiac surgery? Interactive Cardiovascular and Thoracic Surgery. 2014;18:225-229. DOI: https://doi.org/10.1093/icvts/ivt486
  112. 112. Kaleda VI, McCormack DJ, Shipolini AR. Does posterior pericardiotomy reduce the incidence of atrial fibrillation after coronary artery bypass grafting surgery? Interactive Cardiovascular and Thoracic Surgery. 2012;14:384-389. DOI: 10.1093/icvts/ivr099
  113. 113. Ozaydin M, Peker O, Erdogan D, Kapan S, Turker Y, Varol E, Ozguner F, Dogan A, Ibrisim E. N-acetylcysteine for the prevention of postoperative atrial fibrillation: A prospective, randomized, placebo-controlled pilot study. European Heart Journal. 2008;29:625-631. DOI: 10.1093/eurheartj/ehn011
  114. 114. Cavol Cavolli R, Kaya K, Aslan A, Emiroglu O, Erturk S, Korkmaz O, Oguz M, Tasoz R, Ozyurda U. Does sodium nitroprusside decrease the incidence of atrial fibrillation after myocardial revascularization? A pilot study. Circulation. 2008;118:476-481. DOI: 10.1161/CIRCULATIONAHA.107.719377
  115. 115. Fan K, Lee KL, Chiu CS, Lee JW, He GW, Cheung D, Sun MP, Lau CP. Effects of biatrial pacing in prevention of postoperative atrial fibrillation after coronary artery bypass surgery. Circulation. 2000;102:755-760. DOI: https://doi.org/10.1161/01.CIR.102.7.755
  116. 116. Daoud EG, Dabir R, Archambeau M, Morady F, Strickberger SA. Randomized, double-blind trial of simultaneous right and left atrial epicardial pacing for prevention of post-open heart surgery atrial fibrillation. Circulation. 2000;102:761-765. DOI: https://doi.org/10.1161/01.CIR.102.7.761
  117. 117. Levy T, Fotopoulos G, Walker S, Rex S, Octave M, Paul V, Amrani M. Randomized controlled study investigating the effect of biatrial pacing in prevention of atrial fibrillation after coronary artery bypass grafting. Circulation. 2000;102:1382-1387. DOI: https://doi.org/10.1161/01.CIR.102.12.1382
  118. 118. Gerstenfeld EP, Hill MR, French SN, Mehra R, Rofino K, Vander Salm TJ, Mittleman RS. Evaluation of right atrial and biatrial temporary pacing for the prevention of atrial fibrillation after coronary artery bypass surgery. Journal of the American College of Cardiology. 1999;33:1981-1988. DOI: https://doi.org/10.1161/01.CIR.102.12.1382
  119. 119. Chung MK, Augostini RS, Asher CR, Pool DP, Grady TA, Zikri M, Buehner SM, Weinstock M, McCarthy PM. Ineffectiveness and potential proarrhythmia of atrial pacing for atrial fibrillation prevention after coronary artery bypass grafting. The Annals of Thoracic Surgery. 2000;69:1057-1063. DOI: http://dx.doi.org/10.1016/S0003-4975(99)01338-7
  120. 120. Mori SS, Fujii GG, Ishida HH, Tomari SS, Matsuura AA, Yoshida KK. Atrial flutter after coronary artery bypass grafting: Proposed mechanism as illuminated by independent predictors. Annals of Thoracic and Cardiovascular Surgery. 2003;9:50-56 PMID: 12667130
  121. 121. Pap R, Kohári M, Makai A, Bencsik G, Traykov VB, Gallardo R, Klausz G, Zsuzsanna K, Forster T, Sághy L. Surgical technique and the mechanism of atrial tachycardia late after open heart surgery. Journal of Interventional Cardiac Electrophysiology. 2012;35:127-135. DOI: 10.1007/s10840-012-9705-2
  122. 122. Hammon JW, Hines MH. Extracorporeal circulation: Perfusion system. In Cohn LH editor. Cardiac Surgery in the Adult. 4th ed. New York: McGraw-Hill; 2008. pp. 350-370. Ch12
  123. 123. Anné W, van Rensburg H, Adams J, Ector H, Van de Werf F, Heidbüchel H. Ablation of post-surgical intra-atrial reentrant tachycardia. Predilection target sites and mapping approach. European Heart Journal. 2002:23;1609-1616. PMID: 12323161
  124. 124. Nakagawa H, Shah N, Matsudaira K, Overholt E, Chandrasekaran K, Beckman KJ, Spector P, Calame JD, Rao A, Hasdemir C, Otomo K, Wang Z, Lazzara R, Jackman WM. Characterization of reentrant circuit in macroreentrant right atrial tachycardia after surgical repair of congenital heart disease: Isolated channels between scars allow “focal” ablation. Circulation. 2001 Feb 6;103:699-709. DOI: https://doi.org/10.1161/01.CIR.103.5.699
  125. 125. Stewart AM, Greaves K, Bromilow J. Supraventricular tachyarrhythmias and their management in the perioperative period. Continuing Education in Anesthesia, Critical Care and Pain. 2015;15:90-97. DOI: https://doi.org/10.1093/bjaceaccp/mku018
  126. 126. Delacrétaz E. Clinical practice. Supraventricular tachycardia. The New England Journal of Medicine. 2006;354:1039-1051. DOI: 10.1056/NEJMcp051145
  127. 127. Poldermans D, Bax JJ, Boersma E, De Hert S, Eeckhout E, Fowkes G, Gorenek B, Hennerici MG, Iung B, Kelm M, Kjeldsen KP, Kristensen SD, Lopez-Sendon J, Pelosi P, Philippe F, Pierard L, Ponikowski P, Schmid JP, Sellevold OF, Sicari R, Van den Berghe G, Vermassen F, Hoeks SE, Vanhorebeek I, Vahanian A, Auricchio A, Bax JJ, Ceconi C, Dean V, Filippatos G, Funck-Brentano C, Hobbs R, Kearn P, McDonag T, McGregor K, Popescu BA, Reiner Z, Sechtem U, Sirnes PA, Tendera M, Vardas P, Widimsky P, De Caterina R, Agewall S, Al Attar N, Andreotti F, Anker SD, Baron-Esquivias G, Berkenboom G, Chapoutot L, Cifkova R, Faggiano P, Gibbs S, Hansen HS, Iserin L, Israel CW, Kornowski R, Eizagaechevarria NM, Pepi M, Piepoli M, Priebe HJ, Scherer M, Stepinska J, Taggart D, Tubaro M, Task Force for Preoperative Cardiac Risk Assessment and Perioperative Cardiac Management in Non-cardiac Surgery of European Society of Cardiology (ESC), European Society of Anaesthesiology (ESA). Guidelines for pre-operative cardiac risk assessment and perioperative cardiac Management in non-Cardiac Surgery. The task force for preoperative cardiac risk assessment and perioperative cardiac management in non-cardiac surgery of the European Society of Cardiology (ESC) and endorsed by the European Society of Anaesthesiology (ESA). European Journal of Anaesthesiology. 2010;27:92-137. DOI: 10.1097/EJA.0b013e328334c017
  128. 128. Polanczyk CA, Goldman L, Marcantonio ER, Orav EJ, Lee TH. Supraventricular arrhythmia in patients having noncardiac surgery: Clinical correlates and effect on length of stay. Annals of Internal Medicine. 1998;129:279-285. DOI: 10.7326/0003-4819-129-4-199808150-00003
  129. 129. Amar D. Perioperative atrial tachyarrhythmias. Anesthesiology. 2002;97:1618-1623 PMID: 12459693
  130. 130. Amar D, Roistacher N, Burt M, Reinsel RA, Ginsberg RJ, Wilson RS. Clinical and echocardiographic correlates of symptomatic tachydysrhythmias after noncardiac thoracic surgery. Chest. 1995;108:349-354. DOI: https://doi.org/10.1378/chest.108.2.349
  131. 131. Maisel WH, Rawn JD, Stevenson WG. Atrial fibrillation after cardiac surgery. Annals of Internal Medicine. 2001;18(135):1061-1073. DOI: 10.7326/0003-4819-135-12-200112180-00010
  132. 132. Roth A, Elkayam I, Shapira I, Sander J, Malov N, Kehati M, Golovner M. Effectiveness of prehospital synchronous direct-current cardioversion for supraventricular tachyarrhythmias causing unstable hemodynamic states. The American Journal of Cardiology. 2003;91:489-491. DOI: http://dx.doi.org/10.1016/S0002-9149(02)03257-5
  133. 133. Page RL, Joglar JA, Caldwell MA, Calkins H, Conti JB, Deal BJ, Estes NA 3rd, Field ME, Goldberger ZD, Hammill SC, Indik JH, Lindsay BD, Olshansky B, Russo AM, Shen WK, Tracy CM, Al-Khatib SM. 2015 ACC/AHA/HRS guideline for the Management of Adult Patients with Supraventricular Tachycardia: A report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines and the Heart Rhythm Society. Journal of the American College of Cardiology. 2016;67(13):e27-e115. DOI: 10.1016/j.jacc.2015.08.856
  134. 134. Ascione R, Reeves BC, Santo K, Khan N, Angelini G. Predictors of new malignant ventricular arrhythmias after coronary surgery: A case control study. Journal of the American College of Cardiology. 2004;43:1630-1638. DOI: 10.1016/j.jacc.2003.11.056
  135. 135. Huikuri HV, Yli-Mäyry S, Korhonen UR, Airaksinen KE, Ikäheimo MJ, Linnaluoto MK, Takkunen JT. Prevalence and prognostic significance of complex ventricular arrhythmias after coronary arterial bypass graft surgery. International Journal of Cardiology. 1990;27:333-339. DOI: 10.1016/0167-5273(90)90290-L
  136. 136. Smith RC, Leung JM, Keith FM, Merrick S, Mangano DT, Study of Perioperative Ischemia (SPI) Research Group. Ventricular dysrhythmias in patients undergoing coronary artery bypass graft surgery: Incidence, characteristics, and prognostic importance. American Heart Journal. 1992;123:73-81. DOI: https://doi.org/10.1016/0002-8703(92)90749-L
  137. 137. Pinto RP, Romerill DB, Nasser WK, Schier JJ, Surawicz B. Prognosis of patients with frequent premature ventricular complexes and nonsustained ventricular tachycardia after coronary artery bypass graft surgery. Clinical Cardiology. 1996;19:321-324. DOI: 10.1002/clc.4960190408
  138. 138. Topol EJ, Lerman BB, Baughman KL, Platia EV, Griffith LS. De novo refractory ventricular tachyarrhythmias after coronary revascularization. The American Journal of Cardiology. 1986;57:57-59. DOI: http://dx.doi.org/10.1016/0002-9149(86)90951-3
  139. 139. King FG, Addetia AM, Peters SD, Peachey GO. Prophylactic lidocaine for postoperative coronary artery bypass patients, a double-blind, randomized trial. Canadian Journal of Anaesthesia. 1990;37:363-368. DOI: 10.1007/BF03005592
  140. 140. Chung MK. Cardiac surgery: Postoperative arrhythmias. Critical Care Medicine. 2000;28(10):N136-N144 PMID: 11055682
  141. 141. Sapin PM, Woelfel AK, Foster JR. Unexpected ventricular tachyarrhythmias soon after cardiac surgery. The American Journal of Cardiology. 1991;68(10):1099-1100. DOI: http://dx.doi.org/10.1016/0002-9149(91)90505-F
  142. 142. Steinberg JS, Gaur A, Sciacca R, Tan E. New-onset sustained ventricular tachycardia after cardiac surgery. Circulation. 1999;99:903-908. DOI: https://doi.org/10.1161/01.CIR.99.7.903
  143. 143. Yeung-Lai-Wah JA, Qi A, McNeill E, Abel JG, Tung S, Humphries KH, Kerr CR. New-onset sustained ventricular tachycardia and fibrillation early after cardiac operations. The Annals of Thoracic Surgery. 2004;77:2083-2088. DOI: 10.1016/j.athoracsur.2003.12.020
  144. 144. Josephson ME. Recurrent ventricular tachycardia. In: Josephson ME, editor. Clinical Cardiac Electrophysiology: Techniques and Interpretations. 4th ed. Philadelphia: Lippincott Williams &Wilkins; 2008. pp. 446-642
  145. 145. Carmeliet E. Cardiac ionic currents and acute ischemia: From channels to arrhythmias. Physiological Reviews. 1999;79:917-1017 PMID: 10390520
  146. 146. Rubart M, Zipes DP. Mechanisms of sudden cardiac death. The Journal of Clinical Investigation. 2005;115:2305-2315. DOI: 10.1172/JCI26381
  147. 147. Coronel R, Wilms-Schopman FJ, Dekker LR, Janse MJ. Heterogeneities in [K+]o and TQ potential and the inducibility of ventricular fibrillation during acute regional ischemia in the isolated perfused porcine heart. Circulation. 1995;92:120-129. DOI: https://doi.org/10.1161/01.CIR.92.1.120
  148. 148. El-Chami MF, Sawaya FJ, Kilgo P, Stein W 4th, Halkos M, Thourani V, Lattouf OM, Delurgio DB, Guyton RA, Puskas JD, Leon AR.Ventricular arrhythmia after cardiac surgery: Incidence, predictors, and outcomes. Journal of the American College of Cardiology. 2012;60:2664-2671. DOI: 10.1016/j.jacc.2012.08.1011
  149. 149. Miller J, Das M. Differential diagnosis for wide QRS complex tachycardia. In: Zipes D, Jalife J, editors. Cardiac Electrophysiology: From Cell to Bedside. 5th ed. Philadelphia: Elsevier International;2010. P.823-831.
  150. 150. Zipes DP, Camm AJ, Borggrefe M, Buxton AE, Chaitman B, Fromer M, Gregoratos G, Klein G, Moss AJ, Myerburg RJ, Priori SG, Quinones MA, Roden DM, Silka MJ, Tracy C, Priori SG, Blanc JJ, Budaj A, Dean V, Deckers JW, Despres C, Dickstein K, Lekakis J, McGregor K, Metra M, Morais J, Osterspey A, Tamargo JL, Zamorano JL, Smith SC Jr, Jacobs AK, Adams CD, Antman EM, Anderson JL, Hunt SA, Halperin JL, Nishimura R, Ornato JP, Page RL, Riegel B; American College of Cardiology; American Heart Association Task Force; European Society of Cardiology Committee for Practice Guidelines; European Heart Rhythm Association; Heart Rhythm Society. ACC/AHA/ESC 2006 guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: A report of the American College of Cardiology/American Heart Association task force and the European Society of Cardiology Committee for practice guidelines (writing committee to develop guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death) developed in collaboration with the European Heart Rhythm Association and the Heart Rhythm Society. Europace. 2006;8:746-837. DOI: 10.1093/europace/eul108
  151. 151. Benito B, Josephson ME. Rev Esp Cardiol (Engl Ed). 2012;65:939-955. DOI: 10.1016/j.recesp.2012.03.027
  152. 152. Moss AJ, Hall WJ, Cannom DS, Daubert JP, Higgins SL, Klein H, Levine JH, Saksena S, Waldo AL, Wilber D, Brown MW, Heo M. Improved survival with an implanted defibrillator in patients with coronary disease at high risk for ventricular arrhythmia. The New England Journal of Medicine. 1996 Dec;335(26, 26):1933-1940. DOI: 10.1056/NEJM199612263352601
  153. 153. Fogel RI, Prystowsky EN. Management of malignant ventricular arrhythmias and cardiac arrest. Critical Care Medicine. 2000;28:N165-N169 PMID: 11055686
  154. 154. Rousou JA, Engelman RM, Flack JE 3rd, Deaton DW, Owen SG. Emergency cardiopulmonary bypass in the cardiac surgical unit can be a lifesaving measure in postoperative cardiac arrest. Circulation. 1994;90:280-284. PMID: 7955266
  155. 155. Buxton AE, Lee KL, Fisher JD, Josephson ME, Prystowsky EN, Hafley G. A randomized study of the prevention of sudden death in patients with coronary artery disease. The New England Journal of Medicine. 1999;341:1882-1890. DOI: 10.1056/NEJM199912163412503
  156. 156. Moss AJ, Zareba W, Hall WJ, Klein H, Wilber DJ, Cannom DS, Daubert JP, Higgins SL, Brown MW, Andrews ML, for the Multicenter Automatic Defibrillator Implantation Trial II Investigators. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. The New England Journal of Medicine. 2002;346:877-883. DOI: 10.1056/NEJMoa013474
  157. 157. Bardy GH, Lee KL, Mark DB, Poole JE, Packer DL, Boineau R, Domanski M, Troutman C, Anderson J, Johnson G, McNulty SE, Clapp-Channing N, Davidson-Ray LD, Fraulo ES, Fishbein DP, Luceri RM, Ip JH, Sudden Cardiac Death in Heart Failure Trial (SCD-HeFT) Investigators. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. The New England Journal of Medicine. 2005;352:225-237. DOI: 10.1056/NEJMoa043399
  158. 158. Goldenberg I, Moss AJ, McNitt S, Zareba W, Hall WJ, Andrews ML, Wilber DJ, Klein HU, Investigators MADIT-II. Time dependence of defibrillator benefit after coronary revascularization in the multicenter automatic defibrillator implantation trial (MADIT)-II. Journal of the American College of Cardiology. 2006;47:1811-1817. DOI: 10.1016/j.jacc.2005.12.048
  159. 159. Solomon SD, Zelenkofske S, McMurray JJ, Finn PV, Velazquez E, Ertl G, Harsanyi A, Rouleau JL, Maggioni A, Kober L, White H, Van de Werf F, Pieper K, Califf RM, Pfeffer MA, Valsartan in Acute Myocardial Infarction Trial (VALIANT) Investigators. Sudden death in patients with myocardial infarction and left ventricular dysfunction, heart failure, or both. The New England Journal of Medicine. 2005;352:2581-2588. DOI: 10.1056/NEJMoa043938
  160. 160. Zishiri ET, Williams S, Cronin EM, Blackstone EH, Ellis SG, Roselli EE, Smedira NG, Gillinov AM, Glad JA, Tchou PJ, Szymkiewicz SJ, Chung MK. Early risk of mortality after coronary artery revascularization in patients with left ventricular dysfunction and potential role of the wearable cardioverter defibrillator. Circulation. Arrhythmia and Electrophysiology. 2013;6:117-128. DOI: 10.1161/CIRCEP.112.973552
  161. 161. Hohnloser SH, Kuck KH, Dorian P, Roberts RS, Hampton JR, Hatala R, Fain E, Gent M, Connolly SJ, DINAMIT Investigators. Prophylactic use of an implantable cardioverter-defibrillator after acute myocardial infarction. The New England Journal of Medicine. 2004;351:2481-2488. DOI: 10.1056/NEJMoa041489
  162. 162. Steinbeck G, Andresen D, Seidl K, Brachmann J, Hoffmann E, Wojciechowski D, Kornacewicz-Jach Z, Sredniawa B, Lupkovics G, Hofgärtner F, Lubinski A, Rosenqvist M, Habets A, Wegscheider K, Senges J, Investigators IRIS. Defibrillator implantation early after myocardial infarction. The New England Journal of Medicine. 2009;361:1427-1436. DOI: 10.1056/NEJMoa0901889
  163. 163. Morin DP. The Role and Indications of ICD in Patients with Ischemic Cardiomyopathy Undergoing Revascularization [Internet]. 2016. Available from: http://www.acc.org/latest-in-cardiology/articles/2016/03/21/07/42/the-role-and-indications-of-icd-in-patients-with-ischemic-cardiomyopathy-undergoing-revascularization. [Accessed: June 3, 2017]
  164. 164. Bigger JT Jr, for the Coronary Artery Bypass Graft (CABG) Patch Trial Investigators. Prophylactic use of implanted cardiac defibrillators in patients at high risk for ventricular arrhythmias after coronary-artery bypass graft surgery. Coronary artery bypass graft (CABG) patch trial investigators. The New England Journal of Medicine. 1997;337:1569-1575. DOI: 10.1056/NEJM199711273372201
  165. 165. Nageh MF, Kim JJ, Chen LH, Yao JF. Implantable defibrillators for secondary prevention of sudden cardiac death in cardiac surgery patients with perioperative ventricular arrhythmias. Journal of the American Heart Association. 2014;3:e000686. DOI: 10.1161/JAHA.113.000686
  166. 166. Kusumoto FM, Calkins H, Boehmer J, Buxton AE, Chung MK, Gold MR, Hohnloser SH, Indik J, Lee R, Mehra MR, Menon V, Page RL, Shen WK, Slotwiner DJ, Stevenson LW, Varosy PD, Welikovitch L, Heart Rhythm Society, American College of Cardiology, American Heart Association. HRS/ACC/AHA expert consensus statement on the use of implantable Cardioverter-defibrillator therapy in patients who are not included or not well represented in clinical trials. Journal of the American College of Cardiology. 2014;64:1143-1177. DOI: 10.1016/j.jacc.2014.04.008
  167. 167. Moss AJ, Greenberg H, Case RB, Zareba W, Hall WJ, Brown MW, Daubert JP, McNitt S, Andrews ML, Elkin AD, Multicenter Automatic Defibrillator Implantation Trial-II (MADIT-II) Research Group. Long-term clinical course of patients after termination of ventricular tachyarrhythmia by an implanted defibrillator. Circulation. 2004;110:3760-3765. DOI: 10.1161/01.CIR.0000150390.04704.B7
  168. 168. Baerman JM, Kirsh MM, de Buitleir M, Hyatt L, Juni JE, Pitt B, Morady F. Natural history and determinants of conduction defects following coronary artery bypass surgery. The Annals of Thoracic Surgery. 1987;44:150-153 PMID: 3497615
  169. 169. Goldman BS, Hill TJ, Weisel RD, Scully HE, Mickleborough LL, Pym J, Baird RJ. Permanent pacing after open-heart sugery: Acquired heart disease. Pacing and Clinical Electrophysiology. 1984;7:367-371. DOI: 10.1111/j.1540-8159.1984.tb04919.x
  170. 170. Emlein G, Huang SK, Pires LA, Rofino K, Okike ON, Vander Salm TJ. Prolonged bradyarrhythmias after isolated coronary artery bypass graft surgery. American Heart Journal. 1993;126:1084-1090. DOI: 10.1016/0002-8703(93)90658-V
  171. 171. Al-Ghamdi B, Mallawi Y, Shafquat A, Ledesma A, AlRuwaili N, Shoukri M, Khan S, Al Saneia A. Predictors of permanent pacemaker implantation after coronary artery bypass grafting and valve surgery in adult patients in current surgical era. Cardiology Research. 2016;7:123-129. DOI: http://dx.doi.org/10.14740/cr480w
  172. 172. Miriam S, Leonardo B, Solari Gabriel S. Pacemaker following adult cardiac surgery. In: Min M, editor. Cardiac Pacemakers–Biological Aspects, Clinical Applications and Possible Complications. Croatia: InTech; 2011. pp. 135-160. DOI: 10.5772/23402
  173. 173. Raza SS, Li JM, John R, Chen LY, Tholakanahalli VN, Mbai M, Adabag AS. Long-term mortality and pacing outcomes of patients with permanent pacemaker implantation after cardiac surgery. Pacing and Clinical Electrophysiology. 2011;34:331-338. DOI: 10.1111/j.1540-8159.2010.02972.x
  174. 174. Gordon RS, Ivanov J, Cohen G, Ralph-Edwards AL. Permanent cardiac pacing after a cardiac operation: Predictoing the use of permanent pacemakers. The Annals of Thoracic Surgery 1998;66:1698-1704. DOI: http://dx.doi.org/10.1016/S0003-4975(98)00889-3
  175. 175. Limongelli G, Ducceschi V, D'Andrea A, Renzulli A, Sarubbi B, De Feo M, Cerasuolo F, Calabrò R, Cotrufo M. Risk factors for pacemaker implantation following aortic valve replacement: A single centre experience. Heart. 2003;89:901-904. DOI: http://dx.doi.org/10.1136/heart.89.8.901
  176. 176. Erdogan O, Augostini R, Saliba W, Juratli N, Wilkoff BL. Transiliac permanent pacemaker implantation after extraction of infected pectoral pacemaker systems. The American Journal of Cardiology. 1999;84:474-475. DOI: http://dx.doi.org/10.1016/S0002-9149(99)00339-2
  177. 177. Schurr UP, Berli J, Berdajs D, Häusler A, Dzemali O, Emmert M, Seifert B, Genoni M. Incidence and risk factors for pacemaker implantation following aortic valve replacement. Interactive Cardiovascular and Thoracic Surgery. 2010;11:556-560. DOI: 10.1510/icvts.2010.249904
  178. 178. Nardi P, Pellegrino A, Scafuri A, Bellos K, De Propris S, Polisca P, Chiariello L. Permanent pacemaker implantation after isolated aortic valve replacement: Incidence, risk factors and surgical technical aspects. Journal of Cardiovascular Medicine (Hagerstown, Md.). 2010;11:14-19. DOI: 10.2459/JCM.0b013e32832f9fde
  179. 179. Del Rizzo DF, Nishimura S, Lau C, Sever J, Goldman BS. Cardiac pacing following surgery for acquired heart disease. Journal of Cardiac Surgery. 1996;11:332-340. DOI: 10.1111/j.1540-8191.1996.tb00059.x
  180. 180. Sachweh JS, Vazquez-Jimenez JF, Schondube FA, Daebritz SH, Dorge H, Muhler EG, Messmer BJ. Twenty years experience with pediatric pacing: Epicardial and transvenous stimulation. European Journal of Cardio-Thoracic Surgery. 2000;17:455-461. DOI: https://doi.org/10.1016/S1010-7940(00)00364-X
  181. 181. Meimoun P, Zeghdi R, D'Attelis N, Berrebi A, Braunberger E, Deloche A, Fabiani JN, Carpentier A. Frequency, predictors, and consequences of atrioventricular block after mitral valve repair. The American Journal of Cardiology. 2002;89(9):1062-1066. DOI: http://dx.doi.org/10.1016/S0002-9149(02)02276-2
  182. 182. Heinz G, Kratochwill C, Buxbaum P, Laufer G, Kreiner G, Siostrzonek P, Gasic S, Derfler K, Gössinger H. Immediate normalization of profound sinus node dysfunction by aminophylline after cardiac transplantation. The American Journal of Cardiology. 1993;71:346-349. DOI: https://doi.org/10.1016/0002-9149(93)90805-M
  183. 183. Haught WH, Bertolet BD, Conti JB, Curtis AB, Mills RM. Theophylline reverses high-grade atrioventricular block resulting from cardiac transplant rejection. American Heart Journal. 1994;128:1255-1257. DOI: http://dx.doi.org/10.1016/0002-8703(94)90764-1
  184. 184. European Society of Cardiology (ESC), European Heart Rhythm Association (EHRA), Brignole M, Auricchio A, Baron-Esquivias G, Bordachar P, Boriani G, Breithardt OA, Cleland J, Deharo JC, Delgado V, Elliott PM, Gorenek B, Israel CW, Leclercq C, Linde C, Mont L, Padeletti L, Sutton R, Vardas PE. 2013 ESC guidelines on cardiac pacing and cardiac resynchronization therapy: The task force on cardiac pacing and resynchronization therapy of the European Society of Cardiology (ESC). Developed in collaboration with the European heart rhythm association (EHRA). Europace. 2013;15:1070-1118. DOI: 10.1093/europace/eut206

Written By

Bandar Al-Ghamdi

Submitted: 19 March 2017 Reviewed: 18 July 2017 Published: 20 December 2017