Supraventricular Arrhythmias

advertisement
CHAPTER 25
Supraventricular
Arrhythmias
James E. Tisdale
D
rugs may induce a variety of supraventricular
arrhythmias that may be associated with symptoms, hemodynamic instability, stroke, and, in some
cases, an increased risk of death. Supraventricular
arrhythmias that may be induced by drugs include
sinus bradycardia, atrioventricular (AV) nodal blockade, atrial fibrillation or flutter, atrial tachycardia,
and AV nodal reciprocating tachycardia.
SINUS BRADYCARDIA/
ATRIOVENTRICULAR (AV)
NODAL BLOCKADE
Sinus bradycardia is defined as a sinus rate <60
bpm.1 Drugs that inhibit sinus node function
resulting in sinus pauses or sinus arrest will also be
included in this section. Sinus pauses occur when
function of the sinus node is impaired transiently,
leading to “pauses” without p waves on the electrocardiogram (Figure 25–1).1,2 Sinus pauses are also
known as periods of sinus arrest.
AV nodal blockade occurs when conduction of
impulses from the atria to the ventricles through the
AV node is inhibited.3 AV nodal blockade is classified
as first, second, or third degree. First degree AV nodal
blockade is defined as prolongation of the PR interval to >0.2 seconds on an electrocardiogram (ECG).
Second degree AV block is a progression of AV nodal
dysfunction to the point at which some impulses are
not conducted from the atria to the ventricles. Third
degree AV nodal blockade, often referred to as “complete heart block” or “AV dissociation,” is defined by
the absence of a relationship between atrial and ventricular depolarization. During third degree AV nodal
blockade, atrial depolarization occurs independently
of ventricular depolarization, as a result of the complete inability of the AV node to conduct impulses.3
CAUSATIVE AGENTS
Sinus bradycardia with or without sinus pauses
may be associated with a wide variety of drugs
(Table 25–1).4-330 In addition to occurring as a result
22 minutes
23 minutes
FIGURE 25–1. Sinus pauses/sinus
28 minutes
arrest at several time points following ibutilide 1.0 mg/kg in a patient
subsequently determined to have
underlying sinus node dysfunction.
30 minutes
Reproduced with permission from Amin
et al.136
445
446
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–1
Agents Implicated in Drug-Induced Sinus Bradycardia
Drug
Incidence
Level of Evidence
Adenosine4-6
Adrenergic b-receptor blockers7,8
Amiodarone9-11
Baclofen12-14,b
Bupivicaine15,16
Carbamazepine17,18
Cimetidine19-25
Cisplatin26-28
Citalopram29-31
Clonidine32-58
Clozapine59,60
Cocaine61
1–8%
0.6–3.8%
0–3%a
NK
NK
NK
NK
NK
0.1–1.0%
0–17.5%
NK
A
A
NK
4.4–55%
NK
0 –7%
5–16%
0.5–6.7%
0 –4%
0.6%
0.7–2.3%
NK
NK
2.0–13.2%
NK
NKd
B
A
C
B
A
B
B
B
A
C
C
B
B
B
11–24%
0 –4.5%
NK
0–5.5%e
NK
NK
NK
0–13%
OR 2.7 (95% CI, 1.4–5.4)
A
B
C
B
C
C
B
A
B
0–10%
0–0.7%
0.7%
3–29%
5%f
B
B
B
(see page xii for explanation)
Dexmedetomidine62-79
Diazepam21,b
Digoxin80-83
Diltiazem84-96
Dipyridamole97-103,c
Disopyramide104-107
Donepezil108-111
Dronedarone112,113
Escitalopram114
Etidocaine15
Flecainide115-121
Fluoxetine122-128
g-Hydroxybutyric acid129-131
Halothane132-135
Ibutilide136-139
Isradipine140,b
Ketamine141-145
Lidocaine146-151
Methadone152-154
Methylprednisolone155-161
Milrinone162
Neostigmine163-174
Nicardipine175,176
Nitroglycerin177-185
Olanzapine186-189
Paclitaxel190-198
199
Pentazocine
A
C
C
C
C
B
C
A
C
A
B
(Continued)
CHAPTER 25 • Supraventricular Arrhythmias
TABLE 25–1
Agents Implicated in Drug-Induced Sinus Bradycardia
Drug
Propafenone200-203
Propofol204-216
Physostigmine166,217,218
Pyridostigmine166
Ranitidine219-227
Remifentanil228-240
Risperidone241,242
Sevoflurane133,243-252
Sotalol253-274
Succinylcholine275-287
Tacrolimus288,289
Thalidomide290-301
Verapamil132,302-330
447
(Continued)
Incidence
0.7%g
10%h
0–50% (14.7% in analysis
of combined studies)
NK
NK
NK
0–39%
NK
0–53%
2.5–17.2%
0–50%
NK
3.2–53%
0–11%
Level of Evidence
B
B
B
B
C
C
A
C
A
A
A
C
A
A
CI = confidence interval; NK = not known; OR = odds ratio.
a
May be as high as 10% in patients receiving high doses (3 g over 24 hr) of intravenous amiodarone.
b
In overdose.
c
Reported with intravenous use
d
Incidence in g-hydroxybutyric acid intoxication: 38%.130
e
Incidence as high as 21.1% in infants.143
f
Intramuscular administration in children.
g
Oral propafenone.
h
Intravenous propafenone.
of drugs administered orally or intravenously, sinus
bradycardia and sinus pauses may be induced by
drugs administered as eye drops. Numerous cases
of sinus bradycardia associated with topical adrenergic b-receptor antagonists (b-blockers) have been
reported.331,332 The incidence of sinus bradycardia
associated with topical timolol is significantly
higher (18.4%) than that associated with topical
carteolol (4.5%).331 The lower incidence of sinus
bradycardia associated with carteolol may be a
result of the intrinsic sympathomimetic activity of
the drug.
Drugs that have been reported to cause AV
nodal blockade are listed in Table 25–2.4,5,17,83,86,
90,105,107,111,115,190-192,333-447
EPIDEMIOLOGY
The overall incidence of drug-induced sinus bradycardia or AV nodal blockade is unknown. The inci-
dence of sinus bradycardia and sinus pauses associated with specific agents, where known, is presented in Table 25–1, and the incidence of
drug-induced AV nodal blockade associated with
specific drugs, where known, is presented in Table
25–2.
Amiodarone has been reported to cause sinusnode and AV nodal bradyarrhythmias requiring
pacemaker insertion.11 In a study of 15,824
patients with atrial fibrillation and a history of
myocardial infarction who were receiving amiodarone, the incidence of pacemaker implantation
was 2.2% per person-year (mean duration [±SD] of
follow-up, 1.8±1.5 years), and was 5.2% per person-year during the first 90 days of therapy.11 The
odds ratio for pacemaker insertion to treat bradyarrhythmias associated with amiodarone use was
2.14 (95% confidence interval [CI] 1.30 to 3.54).448
Although the incidence of sinus bradycardia associated with conventional doses of intravenous
amiodarone (1 to 2 g/24 hr) is similar to that asso-
448
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–2
Agents Implicated in Drug-Induced Atrioventricular Nodal Blockade
Drug
Incidence
Level of Evidence
(see page xii for explanation)
Aconite333,334
Adenosine4,5,335-351
Adrenergic b-receptor blockers352-361
Amiodarone357,362-366
Amitriptyline367,368
Bupivacaine369,370
Carbamazepine17,371-376
Chloroquine377-379
Cimetidine380
Citalopram381
Clonidine382-385
Cocaine386
Digoxin83,387-402
Diltiazem86,90,359,403-405
Dipyridamole406-408
Disopyramide105,107,409-411
Donepezil111,412
Etomidate413
Flecainide115,414
Guanabenz415
Hydroxychloroquine416
Imipramine417,418
Nicardipine419
Paclitaxel190-192,420
Pentamidine421
Phenylpropanolamine422,423
Propafenone424-426
Propofol427-429
Pyridostigmine430
Sotalol431
Thioridazine432,433
Verapamil359,385,405,434-447
C
A
NK
3–11%
NK
0–14.8%
NK
3.5%
NK
NK
NK
NK
NK
A
A
C
B
C
C
C
C
C
NK
NK
0–2%
NK
0–2%
NK
NK
NK
NK
NK
NK
NK
0 –4.4%
NK
C
B
A
C
B
C
C
C
C
C
C
C
B
C
NK
0.3%
NK
NK
0 –4.2%
NK
0–8%
C
A
C
C
B
C
A
NK = not known.
ciated with oral amiodarone,449 the incidence may
be as high as 10% when doses of 3 g per 24 hours
are administered.450
Sinus bradycardia or arrest or AV nodal blockade
may be caused by digoxin, primarily in overdose or
other situations in which serum digoxin concentrations become elevated.83 The overall incidence of
sinus bradycardia or AV nodal blockade associated
with digoxin is unknown. In an analysis of patients
with a discharge diagnosis of digoxin intoxication,
“definite” sinus bradycardia and sinus pauses <2 seconds were documented in 26% and 19%, respectively, of those with digoxin intoxication.83 In addition,
“definite” second or third degree AV nodal blockade
was documented in 33% of patients with digoxin
intoxication.83 Sotalol has been reported to cause
CHAPTER 25 • Supraventricular Arrhythmias
clinically important sinus bradycardia and sinus
pauses in up to 17.2% of patients being treated for
atrial fibrillation, with 2.5% requiring permanent
pacemaker implantation.269
Zelster et al.359 reviewed a series of 169 consecutive patients admitted or discharged with the
diagnosis of second or third degree AV nodal blockade that was not caused by acute myocardial
infarction, vasovagal syncope, radiofrequency
ablation, or digitalis toxicity. Of these, 54% were
receiving therapy with b-blockers with or without
diltiazem, diltiazem alone, or verapamil. The investigators reported that drug discontinuation was followed by resolution of AV nodal blockade in 41%
of patients, but that 56% of these patients subsequently had recurrence of AV nodal blockade in
the absence of causative drugs. Based on their criteria for causation, the investigators concluded
that drugs were the specific cause of AV nodal
blockade in only 15% of the patients who were
receiving therapy with b-blockers with or without
diltiazem, diltiazem alone, or verapamil.359
MECHANISMS
Drug-induced sinus bradycardia may be caused by
inhibition of automaticity of the node, slowing of
sinus node conduction, or prolongation of sinus
node repolarization (Table 25–3). AV nodal blockade may be caused by agents that inhibit AV node
conduction or prolong AV node repolarization
(Table 25–4). Both the sinus node and the AV node
are heavily influenced by sympathetic and
parasympathetic nervous system activity. Drugs
that inhibit activity of the sympathetic nervous
TABLE 25–3
449
system (e.g., b-blockers) and drugs that stimulate
the parasympathetic nervous system (e.g., neostigmine, physostigmine, pyridostigmine) may cause
sinus bradycardia or AV nodal blockade. The action
potentials of both the sinus node and the AV node
depend primarily on calcium and sodium flux, and
calcium or sodium channel inhibitors may cause
sinus bradycardia, AV nodal blockade, or both.
Nitrates may cause a syndrome associated with
sinus bradycardia and hypotension.177-185 Sinus
bradycardia induced by nitrates is an idiosyncratic
reaction. This syndrome resembles neurocardiogenic syncope, but may occur when patients are in
the supine position. The mechanism of nitrateinduced sinus bradycardia remains unclear, but
evidence suggests that nitrates may stimulate
peripheral sensory receptors with vagal afferents to
the medulla, resulting in sympathetic nervous system inhibition via stimulation of central a2-receptors (Table 25–3).451
CLINICAL PRESENTATION AND
DIFFERENTIAL DIAGNOSIS
Normal heart rate is usually defined somewhat
arbitrarily as ranging between 60 and 100 bpm.
However, many individuals routinely have heart
rates <60 bpm and even <50 bpm without evidence
of symptoms. Patients with symptomatic sinus
bradycardia resulting in the need for medical attention typically have heart rates between 30 and 50
bpm, resulting in hemodynamic compromise.
Signs and symptoms associated with clinically
important sinus bradycardia and sinus pauses are
listed in Table 25–5.1
Mechanisms of Drug-Induced Sinus Bradycardia
Drug
All drugs that may cause sinus bradycardia (except nitrates)
Adrenergic b-receptor blockers
Mechanism
Inhibition of automaticity of the sinus node and/or slowing
of sinus node conduction and/or prolongation of
sinus node repolarization
Inhibit activity of sympathetic nervous system, leading to
inhibition of automaticity of sinus node
Clonidine
Stimulates central a2-receptors, reducing release of
norepinephrine
Neostigmine, physostigmine, pyridostigmine
Stimulate activity of the parasympathetic nervous system,
leading to inhibition of automaticity of sinus node
Stimulates peripheral sensory receptors with vagal afferents
to the medulla, resulting in sympathetic nervous system
inhibition via stimulation of central a2-receptors
Nitroglycerin
450
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–4
Mechanisms of Drug-Induced Atrioventricular (AV) Nodal Blockade
Drug
All drugs that may cause AV nodal blockade
Adrenergic b-receptor blockers
Mechanism
Inhibition of conduction through the AV node
Inhibit activity of sympathetic nervous system, leading to
inhibition of conduction through the AV node
Clonidine
Stimulates central a2-receptors, reducing release of norepinephrine
Stimulate activity of the parasympathetic nervous system,
leading to inhibition of conduction through the AV node
Neostigmine, physostigmine, pyridostigmine
First degree AV nodal block is a common electrocardiographic phenomenon that is almost
always asymptomatic3, and is therefore not a druginduced disease as defined in this text. However,
second and third degree AV blockade can result in
bradycardia, leading to symptoms that result in the
need for medical attention or even requiring hospitalization.
Second degree AV block occurs most commonly in two distinct patterns. Mobitz type I seconddegree AV block (also known as Wenckebach) is
characterized by progressive prolongation of the
PR interval on ECG until a P wave is not followed
by a QRS complex (Figure 25–2).3 Mobitz type I is
usually caused by impulse conduction delay in the
AV node (as opposed to further down in the bundle of His).1 Mobitz type II second degree AV block
is typified by constant PR intervals with abrupt,
intermittent absence of QRS complexes (Figure
25–3). Mobitz type II second degree AV block often
occurs in specific patterns with P wave to QRS complex ratios such as 2:1, 3:1, or 4:1. This type of second degree AV block is likely caused by
impulse-conduction delay in the bundle of His.
During third degree AV nodal blockade, the AV
node is completely unable to conduct impulses,
and atrial depolarization occurs independently of
ventricular depolarization. Therefore, P waves and
QRS complexes are present on ECG but bear no
relationship to one another (Figure 25–4). Third
degree AV nodal blockade can occur either in the
AV node itself or further down in the bundle of
His.3
Second or third degree AV nodal blockade may
result in bradycardia, with heart rates in the range
of 40-60 bpm. Symptoms of second or third degree
AV nodal blockade are the same as those associated
with sinus bradycardia (Table 25–5).
Drug-induced sinus bradycardia or AV nodal
blockade must be distinguished from other causes
of sinus or AV node impairment. Dysfunction of
the sinus node unrelated to drug therapy is common, with a prevalence as high as 1 in 600 patients
over the age of 65 years.452 Intrinsic AV nodal
abnormalities also may occur during the aging
process. Transient sinus node dysfunction may
occur in the setting of acute myocardial ischemia
or infarction when the infarct/ischemia-related
vessel is the right coronary artery or the left circumflex artery, but permanent sinus-node damage
in this setting is uncommon. Myocardial infarction
may result in some degree of permanent AV nodal
dysfunction in some patients. Conditions to consider in the differential diagnosis of drug-induced
sinus bradycardia or AV nodal blockade are listed
in Table 25–6.1
TABLE 25–5 Signs and Symptoms Associated
with Drug-Induced Sinus Bradycardia and
Atrioventricular Nodal Blockade
•
•
•
•
•
•
•
Dizziness
Light-headedness
Fatigue
Weakness
Syncope
Chest pain
Symptoms of heart failure
FIGURE 25–2. Mobitz type I second degree AV nodal
blockade associated with phenylpropanolamine.
Reproduced with permission from Woo et al.423
CHAPTER 25 • Supraventricular Arrhythmias
1
2
3
451
ischemia or infarction. However, it may not be possible or desirable in every case to discontinue therapy with drugs suspected of causing sinus
bradycardia or AV nodal blockade. For example, bblocker therapy should not generally be discontinued in a patient being treated for heart failure or
after myocardial infarction. Rather, therapy with
the b-blocker should be continued and implantation of a permanent pacemaker considered as a
means to protect against possible drug-induced
sinus or AV node dysfunction.
RISK FACTORS
FIGURE 25–3. Mobitz type II second degree AV nodal
blockade associated with amiodarone.
Reproduced with permission from Mangiardi et al.363
Drug-induced sinus or AV node dysfunction
can often be distinguished from non–drug-related
sinus bradycardia or AV nodal blockade. When a
patient with symptoms that appear to be related to
sinus bradycardia/sinus pauses/sinus arrest or AV
nodal inhibition seeks medical attention, therapy
with drugs known to induce sinus or AV node dysfunction should typically be discontinued. If the
sinus or AV node dysfunction persists after five
half-lives of the respective drug(s) have passed, a
drug-induced cause may be ruled out. While awaiting drug washout, evaluation and diagnostic testing for nonpharmacologic causes can be
performed, including determination of serum
potassium and magnesium concentrations, thyroid
function tests, and assessment for myocardial
Risk factors for drug-induced sinus bradycardia are
listed in Table 25–7.41,136,453 Conditions that may
result in elevated plasma concentrations of drugs
known to cause sinus bradycardia increase the risk.
Patients with kidney disease who are receiving renally eliminated drugs that may cause sinus bradycardia (atenolol, clonidine, digoxin) are also at
increased risk. Kidney disease has been identified as
a risk factor for sinus bradycardia associated with
clonidine.41 Drugs that have been reported to cause
sinus bradycardia that require dose adjustment in
patients with kidney disease are listed in Table 25–8.
In addition, concomitant use of drugs that inhibit
the hepatic metabolism of drugs known to induce
sinus bradycardia should be avoided (Table 25–9).454
Other interactions may result in sinus bradycardia. Dipyridamole inhibits the cellular uptake of
adenosine455; therefore, patients receiving adenosine concomitantly with dipyridamole are at
increased risk of adenosine-associated sinus brady-
FIGURE 25–4. Third degree
AV nodal blockade associated
with carbamazepine.
Reproduced with permission
from Ide and Kamijo.376
452
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–6 Conditions to Consider in the
Differential Diagnosis of Drug-Induced Sinus
Bradycardia or Atrioventricular Nodal Blockade1
TABLE 25–7 Risk Factors for Drug-Induced Sinus
Bradycardia or Atrioventricular (AV) Nodal
Blockade
Intrinsic causes
• Idiopathic degeneration due to aging
• Myocardial ischemia/infarction (transient)
• Infiltrative diseases (sarcoidosis, amyloidosis,
hemochromatosis)
• Collagen vascular diseases (systemic lupus erythematosus, scleroderma, rheumatoid arthritis)
• Myotonic muscular dystrophy
• Surgical trauma (valve replacement, heart transplantation, correction of congenital heart disease)
• Infectious diseases (Chagas’ disease, endocarditis)
Extrinsic causes
• Autonomically mediated syndromes
• Neurocardiac syncope
• Carotid sinus hypersensitivity
• Situational disturbances (coughing, micturition,
defecation, vomiting)
• Hypothyroidism
• Hypothermia
• Electrolyte abnormalities
• Hyperkalemia
• Hypokalemia
• Hypermagnesemia
Sinus bradycardia41,136,453
• Pretreatment heart rate <60 bpm
• Underlying sinus-node dysfunction (may occur with
advancing age)
• Impaired baroreflex control (risk factor for nitroglycerin-induced sinus bradycardia)
• Concomitant use of >1 sinus-node inhibiting drug
• Elevated plasma drug concentrations due to organ
dysfunction or drug interactions
Atrioventricular nodal blockade
• Concomitant use of >1 AV nodal blocking drug
• Pretreatment PR interval >0.2 sec
• Underlying AV nodal disease (may occur with
advancing age)
• Elevated plasma drug concentrations due to organ
dysfunction or drug interactions
• Hypothyroidism (may be a risk factor for amiodarone-induced AV block)363
cardia/sinus pauses.6 A number of drugs inhibit the
elimination of digoxin, particularly amiodarone,456
verapamil,457 and quinidine,458 and concomitant
use of these drugs with digoxin increases the risk of
digoxin-induced sinus bradycardia, unless appropriate digoxin dose reduction is implemented. In
addition, patients receiving therapy with more
than one drug known to reduce heart rate are at
increased risk.41,459
Women are at greater risk for amiodaroneinduced bradycardia requiring implantation of a
permanent pacemaker.460 In a study of 1,005
patients with new-onset atrial fibrillation, the hazard ratio for amiodarone use requiring a permanent pacemaker was 4.69 (95% CI, 1.99 to 11.05)
in women, as compared with 1.05 (95% CI, 0.42 to
2.58) in men.460 Mechanisms for the increased risk
of bradycardia requiring pacemaker implantation
in women are unknown.
Specific risk factors for the majority of druginduced AV nodal blockade have not been identified. In the study by Zelster et al.359 of 169
consecutive cases of AV nodal blockade, there were
no significant differences in age, male sex, or presence of hypertension or ischemic heart disease
between patients receiving therapy with b-blockers
with or without diltiazem or verapamil and those
not receiving therapy with drugs that could induce
AV nodal blockade. Possible risk factors for druginduced AV nodal blockade are listed in Table 25–7.
Risk factors have been identified for AV nodal blockade induced by digoxin, and include the drug interactions listed above, inadequate dose reduction in
patients with kidney disease, hypokalemia, hypomagnesemia, hypoxia, and hypothyroidism.461
MORBIDITY AND MORTALITY
The incidence of morbidity, hospitalization, or
death associated with drug-induced sinus bradycardia or AV nodal blockade is unknown. Druginduced sinus bradycardia or AV nodal blockade
may result in the need for temporary or permanent
pacemaker implantation.11,30,448 In a population of
patients with new-onset atrial fibrillation, the
adjusted hazard ratio for amiodarone use leading
to a requirement for a permanent pacemaker was
2.01 (95% CI, 1.08 to 3.76).460 Death due to druginduced sinus bradycardia or AV block is likely very
uncommon.
PREVENTION
Drug-induced sinus or AV node dysfunction (or
both) is often preventable (Table 25–10). When
CHAPTER 25 • Supraventricular Arrhythmias
TABLE 25–8 Drugs Known to Cause Sinus
Bradycardia or Atrioventricular Nodal Blockade
That May Require Dose Adjustment in Patients
with Kidney Disease
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
•
Acebutolol
Atenolol
Baclofen
Betaxolol
Bisoprolol
Carteolol
Cimetidine
Cisplatin (recommended to be avoided in cases of
preexisting renal dysfunction)
Clonidine
Dexmedetomidine
Digoxin
Disopyramide
Flecainide
Guanabenz
Isradipine
Nadolol
Nicardipine
Ranitidine
Sotalol
possible, drugs with the potential to inhibit sinus
or AV node function should be avoided in patients
with known sinus or AV node dysfunction in the
absence of a functioning pacemaker. Once a pacemaker has been implanted, however, patients with
sinus node dysfunction may receive sinus
node–inhibiting drugs, and patients with AV node
dysfunction may receive AV node–inhibiting
453
agents. Patients who are taking drugs with the
potential to cause sinus bradycardia, sinus pauses,
or AV nodal blockade should be taught to monitor
their heart rate daily, and to consult their pharmacist or physician if heart rate falls below 50 bpm or
if they experience symptoms of bradycardia (Table
25–5). Combinations of drugs that may inhibit
sinus or AV node function should be minimized,
but are not contraindicated if the benefits of such
therapy outweigh the risks.
Drug-induced sinus or AV node dysfunction
may be prevented by not exceeding maximum
daily doses of drugs known to cause these druginduced diseases and by appropriate dose adjustment of specific drugs to account for organ
dysfunction, drug interactions, or both.
Maintaining serum digoxin concentrations <2.0
ng/mL may prevent digoxin-associated sinus or AV
node dysfunction. In certain circumstances, substituting medications within a class may prevent
drug-induced sinus or AV node dysfunction. For
example, a patient with kidney disease who is
receiving atenolol is at higher risk for atenololinduced sinus bradycardia or AV nodal blockade as
a result of diminished atenolol clearance. In this
situation, replacing atenolol with a non–renally
cleared b-blocker such as metoprolol or propanolol
may reduce the likelihood of drug-induced sinus
bradycardia or AV nodal blockade.
Patients receiving drugs known to inhibit AV
nodal conduction should undergo a 12-lead ECG
periodically and at least every 6 months. First
degree AV block is not an absolute contraindication
to receiving AV nodal blocking drugs, but the PR
interval should be monitored every 3 to 6 months
TABLE 25–9 Drugs Known to Cause Sinus Bradycardia or Atrioventricular Nodal Blockade That Are
Substrates for Enzymes of the Cytochrome P-450 System454
1A2
Amitriptyline
Clozapine
Olanzapine
Propranolol
Verapamil
2B6
Methadone
2C8
Paclitaxel
2C19
Amitriptyline
Citalopram
Diazepam
Propranolol
2C9
Amitriptyline
Fluoxetine
2D6
Amitriptyline
Flecainide
Carvedilol
Fluoxetine
Imipramine
Lidocaine
Metoprolol
Nebivolol
Propafenone
Propranolol
Thioridazine
Timolol
3A4
Amiodarone
Cocaine
Carbamazepine
Diazepam
Diltiazem
Dronedarone
Lidocaine
Methadone
Nicardipine
Paclitaxel
Tacrolimus
Verapamil
454
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–10 Approaches to Help Prevent DrugInduced Sinus Bradycardia and Atrioventricular
(AV) Nodal Blockade
TABLE 25–11 Management of Drug-Induced
Sinus Bradycardia or Atrioventricular (AV) Nodal
Blockade
For all drugs that may cause sinus bradycardia, AV
block, or both
• Patient should take pulse daily, report if <50 bpm.
• Do not exceed maximum daily doses.
• Use combinations of drugs that may cause sinus
bradycardia or AV block only when necessary and
when the benefits likely outweigh the risks.
• Avoid sinus or AV node–inhibiting drugs in patients
with underlying sinus node dysfunction, unless a
functioning pacemaker is present (in the case of AV
node inhibiting drugs, there must be a functioning
ventricular pacemaker).
For digoxin
• Measure serum digoxin concentrations:
• If kidney function is changing.
• If a drug that interacts with digoxin is added to
therapy.
• Every 6 mo if there is no organ dysfunction or concomitant use of interacting drugs.
• Discontinue the causative agent
• Temporary pacemaker
• If underlying sinus or AV node dysfunction—permanent pacemaker may be necessary (if AV node dysfunction, must be a ventricular pacemaker)
• In severe cases: atropine 0.5–1.0 mg every 3–5 minutes until heart rate increases or total dose of 0.04
mg/kg is administered
• If due to calcium-channel blocker or b-blocker overdose:
• Gastric lavage
• Activated charcoal 25–50 g in an aqueous slurry of
120–240 mL water
• Glucagon 2.0–5.0 mg intravenously
• Temporary pacemaker, if necessary
to ensure that AV nodal blockade is not progressing.
In addition, combinations of AV nodal blocking
drugs are best avoided in patients with pretreatment PR intervals >0.2 seconds. If the PR interval
progresses to >0.2 seconds while on AV nodal blocking therapy, it is not necessary to discontinue therapy, but more frequent ECG monitoring is
recommended and administration of additional AV
nodal blocking agents should be avoided.
MANAGEMENT
Management options for patients with druginduced sinus bradycardia or AV nodal blockade are
presented in Table 25–11. In some cases, a reduction
in dose of the offending medication may be sufficient, but in most cases in which drug-induced sinus
or AV node dysfunction has resulted in a hospitalization, discontinuation of therapy is necessary. In
the case of sinus bradycardia or AV block induced by
adenosine, treatment is not usually required because
the drug is metabolized very quickly (half–life 5 10
seconds) and the heart rate usually returns to pretreatment values within 20 to 30 seconds.
Percutaneous insertion of a temporary pacemaker may be necessary. If the patient is subsequently
diagnosed with underlying intrinsic sinus or AV node
dysfunction, therapy with the offending medication
may be reinitiated after the implantation of a permanent pacemaker. In severe cases, intravenous atropine
may be administered in doses of 0.5 to 1.0 mg every
3 to 5 minutes to a total dose of 0.04 mg/kg, the dose
at which full blockade of the parasympathetic nervous system occurs in humans.462
If the drug-induced bradycardia or AV block is
a result of an overdose of verapamil, diltiazem or a
b-blocker, gastric lavage may be performed if the
patient presents within 1 hour of ingestion.
Activated charcoal may be administered and may
be particularly useful if the drug was ingested in
the form of a sustained-release preparation.
Although administration of calcium salts has been
shown to be effective for reversing the hemodynamic effects of calcium-channel blockers, calcium
salts have not been particularly effective for reversing the electrophysiologic effects of these drugs.463
Glucagon 2 to 5 mg intravenously may be effective
for reversing the bradycardic effects of calciumchannel blockers or a b-blocker.463,464 Insertion of a
temporary pacemaker may be necessary.
For patients with a history of myocardial infarction or heart failure in whom sinus bradycardia or
AV nodal blockade induced by b-blockers develops,
implantation of a permanent pacemaker may be
necessary to allow the patient to continue therapy
with these agents,465 which have been shown to
prolong survival in these specific conditions.466,467
INFORMATION FOR PATIENTS
Patients should be instructed that specific drug(s)
may cause the heart rate (pulse) to become slower.
Patients should be taught to take their pulse and
to monitor their heart rate daily. Patients should
be instructed to consult their pharmacist or physi-
CHAPTER 25 • Supraventricular Arrhythmias
cian if their heart rate falls below 50 beats per
minute, or if they feel lightheaded, dizzy, tired,
weak, short of breath, or experience chest pain.
ATRIAL FIBRILLATION/
ATRIAL FLUTTER
Atrial fibrillation is a supraventricular arrhythmia that is characterized on ECG by an irregularly irregular pattern of narrow QRS complexes, an
absence of discernible P waves, and an undulating baseline (Figure 25–5). Atrial flutter is a
supraventricular arrhythmia that is characterized
on ECG by a regular pattern of narrow QRS complexes, with discernible P waves that exhibit a
“sawtooth” appearance (Figure 25–6). Atrial flutter is often associated with a ratio of P waves to
QRS complexes of 4:1, 3:1 or, in some cases, 2:1.
CAUSATIVE AGENTS
Drugs that have been associated with atrial fibrillation or atrial flutter are listed in Table
25–12.123,162,196,218,349,468-545 The majority of drugs
that have been associated with induction of atrial
fibrillation or flutter are cardiovascular agents, but
drugs from other classes have been implicated as
well, including the bisphosphonate drugs alendronate496,497 and zoledronic acid.545
EPIDEMIOLOGY
The overall incidence of drug-induced atrial fibrillation or flutter is not known, but it is likely very low.
Adenosine has been reported to cause a substantial
incidence of atrial fibrillation in patients undergoing treatment for AV nodal reentrant tachycardia.475
Although the incidence of alcohol-induced atrial
fibrillation (often referred to as the “holiday heart”
syndrome) is unknown, in a case–control study of
455
patients with acute idiopathic atrial fibrillation, 62%
of cases were associated with heavy alcohol use.489
In a prospective cohort study of the association
between self-reported alcohol use and incident atrial fibrillation, moderate alcohol intake was not associated with new-onset atrial fibrillation. However,
consumption of 35 or more drinks per week was
associated with a hazard ratio for atrial fibrillation of
1.45 (95% CI, 1.02 to 2.04) in men.546 About 5% of
cases of atrial fibrillation in men were estimated to
be attributable to heavy alcohol intake.
In a randomized study of the prevention of fractures in postmenopausal women, the bisphosphonate drug alendronate was found to increase the risk
of “serious” atrial fibrillation (1.5% vs. 1.0% in the
placebo group; relative hazard, 1.51; 95% CI, 0.97 to
2.40; P = 0.07).496 However, alendronate did not
increase the risk of “all atrial fibrillation adverse
events.” In addition, in a case–control study in
women, a higher proportion of patients with atrial
fibrillation had ever used alendronate as compared
with those who had never used any bisphosphonate
(odds ratio of atrial fibrillation, 1.86; 95% CI, 1.09 to
3.15).497 Furthermore, another bisphosphonate drug,
zoledronic acid, was reported to increase the risk of
“serious” atrial fibrillation as compared with placebo
(1.3% vs. 0.5%, P<0.001).545 However, zoledronic acid
did not increase the risk of all atrial fibrillation events.
In a cohort study of 15,795 patients with fractures
treated with oral bisphosphonates, the adjusted risk
of atrial fibrillation was higher than that in an ageand sex-matched group of patients with fractures
who did not receive the drugs (1.18; 95% CI, 1.08 to
1.29).547 A database analysis of over 40,000 women
conducted in the United Kingdom showed no evidence of an overall risk of bisphosphonate-associated
atrial fibrillation, but a potential increased risk of atrial fibrillation associated with alendronate during the
first few months of therapy.548 However, other studies
have not reported an increased risk of atrial fibrillation associated with bisphosphonates.549,550
Determining whether bisphosphonate drugs are associated with atrial fibrillation requires further study.
FIGURE 25–5. Atrial
fibrillation induced by
albuterol.
Reproduced with permission from Breeden and
Safirstein.485
456
SECTION VI • Drug-Induced Cardiovascular Diseases
FIGURE 25–6. Atrial flutter with 2:1 atrioventricular conduction induced by propafenone.
Reproduced with permission from Tai et al.498
New-onset atrial flutter associated with drugs is
also uncommon, but has been reported to occur in
patients with atrial fibrillation who are receiving
Vaughan Williams class IC antiarrhythmic agents (flecainide or propafenone) or amiodarone (Table 25–12).
MECHANISMS
Mechanisms by which some drugs induce atrial fibrillation or flutter are presented in Table 25–13. Atrial
fibrillation is believed to be induced by ectopic
impulses originating from pulmonary veins, atria, or
both, and it has been proposed that adenosine may
induce increased atrial ectopic activity, which may
then trigger the development of atrial fibrillation.475
In addition, atrial fibrillation is maintained by multiple reentrant wavelets, and evidence indicates that
the shorter the wavelength, the more likely that atrial fibrillation develops and is sustained.551,552
Wavelength is the product of the atrial conduction
velocity and the atrial effective refractory period.551,552 Adenosine has been shown to shorten the
atrial effective refractory period, and therefore may
promote reduction in atrial wavelength and the
development of multiple reentrant atrial
wavelets.553,554 The mechanism of alcohol-induced
atrial fibrillation is believed to be related to increased
sympathetic nervous system stimulation.494
Mechanisms by which the bisphosphonate
drugs alendronate and zoledronic acid may cause
atrial fibrillation are unclear. It has been suggested
that bisphosphonate drugs may cause atrial fibrillation through the release of inflammatory
cytokines,496,555 which have been associated with
the development of atrial fibrillation.556 Further
study is necessary to determine the mechanism of
bisphosphonate-induced atrial fibrillation and to
confirm that this is truly a disease that is induced
by drugs from this class.557
Many of the drugs that have been reported to
cause atrial fibrillation or atrial flutter have commonly been used for the management of these arrhythmias. Despite the fact that flecainide has been shown
to be effective for the prevention and management of
atrial fibrillation, data from studies in animals indicate that flecainide may shorten atrial conduction
velocity558 and wavelength,559 effects which may promote the development of atrial fibrillation.
Amiodarone-induced atrial fibrillation may, at least in
some cases, be due to amiodarone-induced thyrotoxicosis.560.561 Dobutamine has been shown to shorten
atrial effective refractory periods,562 while theophylline has been shown to increase atrial automaticity.563 Mechanisms by which other drugs may induce
atrial fibrillation or flutter require further study.
CLINICAL PRESENTATION AND
DIFFERENTIAL DIAGNOSIS
The symptoms of drug-induced atrial fibrillation
and atrial flutter are related to the degree of tachycardia and the resultant effect on blood pressure
and cardiac output (Table 25–14). The symptoms of
atrial fibrillation may be indistinguishable from
those of atrial flutter, and therefore atrial fibrillation and atrial flutter must be distinguished from
CHAPTER 25 • Supraventricular Arrhythmias
TABLE 25–12
457
Agents Implicated in Drug-Induced Atrial Fibrillation or Atrial Flutter
Drug
Incidence
Level of Evidence
(see page xii for explanation)
Adenosine 349,468-482
Albuterol483-485
Alcohol486-495
Alendronate496,497
Amiodarone498, 499
Caffeine500
Diltiazem501
Dobutamine162,502-516
Enoximone513
Flecainide517,518
Fluoxetine123,519
Ipratropium bromide520
Levosimendan515
Loxapine519
Metaproterenol521
Methylprednisolone522-526
Milrinone162,527-530
Mitoxantrone531
Ondansetron532,533
Paclitaxel196,534
Physostigmine218,535
Propafenone498,518,536
Theophylline483,537-542
Verapamil543,544
Zoledronic acid545
1–12%a
NK
NK
0.5%
OR, 1.86 (95% CI, 1.09–3.15)
NK
NKb
NK
0–18%
8.3%
NK
B
C
B
A
B
B
C
A
A
C
NK
NK
0–9.1%
NK
2.5%
NK
2.9–5%
1.4%
NK
2% (atrial flutter)
1–1.7% (atrial fibrillation)
NK
9%c
NK
C
C
A
C
A
C
A
B
C
B
5%
0.8%
C
A
C
C
B
CI = confidence interval; NK = not known; OR = odds ratio.
a
Atrial fibrillation, 1–11%, atrial flutter, 1%.
b
Espresso only.
c
Percent of patients with atrial fibrillation who develop new atrial flutter during propafenone therapy.536
one another (and from other tachyarrhythmias) by
ECG (Figures 25–5 and 25–6).
The onset of drug-induced atrial fibrillation or
flutter is variable, depending on the inducing drug.
Adenosine-induced atrial fibrillation or flutter occurs
within 1 minute of administration of this extremely
short-acting agent.349 In a study of amiodarone- or
propafenone-induced atrial flutter, the mean (±SD)
time of onset was 5.0+5.5 months after the initiation
of treatment,498 whereas atrial flutter has been
reported in association with flecainide therapy of 2
months’ duration.517 The duration of episodes of
drug-induced atrial fibrillation or flutter is also somewhat variable; a case of albuterol-induced atrial fibrillation lasted several hours,484 whereas atrial flutter
associated with amiodarone or propafenone may
require intervention to terminate the arrhythmia.498
Sinus rhythm was the preceding rhythm in 88%
of patients in whom atrial fibrillation associated with
adenosine developed, whereas the remaining 12% of
patients had atrial ectopic activity prior to the development of atrial fibrillation.475 The mean ventricular
rate associated with adenosine-induced atrial fibrillation was 107+43 bpm.475 Adenosine-induced
458
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–13
Mechanisms of Drug-Induced Atrial Fibrillation/Flutter
Drug
Adenosine, dobutamine, flecainide
Alcohol
Theophylline
Other drugs
TABLE 25–14 Signs and Symptoms Associated
with Drug-Induced Atrial Fibrillation/Flutter
•
•
•
•
•
Palpitations
Dizziness
Light-headedness
Shortness of breath
Chest pain (if underlying coronary artery disease is
present)
• Near-syncope
• Syncope
episodes of atrial fibrillation tend to be of short duration. The mean duration of episodes of atrial fibrillation associated with adenosine was 5.6+6.7 minutes
(range, 8 seconds to 20.7 minutes).475
Conditions to consider in the differential diagnosis of drug-induced atrial fibrillation/flutter are
presented in Table 25–15. Non–drug-induced atrial
fibrillation/flutter must be considered; drug-induced
atrial fibrillation/flutter is more likely in patients
who are receiving drugs that have been reported to
cause atrial fibrillation or flutter, particularly in
patients with no known risk factors or causes for
non–drug-induced atrial fibrillation/flutter, such as
hypertension, ischemic heart disease, heart failure,
valvular heart disease, rheumatic fever, or hyperthyroidism, or in those who have undergone thoracic
surgery within the previous 2 to 5 days.
Mechanism
Shorten atrial effective refractory period/atrial wavelength
Sympathetic nervous system stimulation
Increases atrial automaticity
Unknown
TABLE 25–15 Conditions to Consider in the
Differential Diagnosis of Drug-Induced Atrial
Fibrillation/Flutter
•
•
•
•
•
Atrial fibrillation or flutter not induced by drugs
Sinus tachycardia
Atrial tachycardia
Atrioventricular nodal reentrant tachycardia
Ventricular tachycardia
(58%), and therefore may be somewhat predictive
of impending drug-induced atrial fibrillation.475
Alcohol dose is a risk factor for alcohol-induced
atrial fibrillation. In one study, the mean dose of
alcohol consumed during the week prior to the
atrial fibrillation episode was 186g, as compared
with 86g in a control population in whom alcoholinduced atrial fibrillation did not develop.493
Another study found that the risk of alcoholinduced atrial fibrillation was increased in patients
who consumed more than an average of 30g alcohol daily.492 Alcohol withdrawal may increase the
risk of alcohol-induced atrial fibrillation.490
In patients receiving amiodarone for treatment
of atrial fibrillation, amiodarone-induced atrial flutter does not appear to be related to age, sex, presence
of structural heart disease, left ventricular function,
or duration of atrial fibrillation; however, left atrial
enlargement (>40 mm) may be a risk factor.498
Further study is needed to identify risk factors for
drug-induced atrial fibrillation or atrial flutter.
RISK FACTORS
Although all risk factors for drug-induced atrial fibrillation or flutter have not yet been fully characterized, known risk factors are presented in Table
25–16. Atrial fibrillation associated with adenosine
does not appear to be related to sex or age and may
occur in patients with no history of atrial fibrillation.475 Atrial fibrillation induced by adenosine is
not related to the type of arrhythmia being treated.475 Premature atrial contractions occur significantly more frequently in patients who have
experienced adenosine-induced atrial fibrillation
(100%) as compared with those who did not
MORBIDITY & MORTALITY
Symptoms associated with drug-induced atrial fibrillation or flutter could potentially result in hospitalization or prolonged duration of stay in a
hospital or critical care unit, although prolonged
duration of hospital stay as a result of druginduced atrial fibrillation or flutter has not been
reported. Hypothetically, drug-induced atrial fibrillation could result in stroke, but this has not yet
been described. Death due to drug-induced atrial
fibrillation or flutter has not been reported.
CHAPTER 25 • Supraventricular Arrhythmias
459
TABLE 25–16 Risk Factors for Drug-Induced
Atrial Fibrillation/Flutter
TABLE 25–17 Approaches to Help Prevent DrugInduced Atrial Fibrillation/Flutter
Adenosine
• Premature atrial contractions
Alcohol
• Dose > 30g daily
• Withdrawal
Amiodarone
• Left atrial hypertrophy
Other drugs
• Unknown
Alcohol492,493
• Avoid binge drinking/excessive doses
• Consume <30g daily
Methylprednisolone524
• Propafenone 300 mg three times daily
Other drugs
• Administer lowest effective dose
PREVENTION
For the most part, specific methods of prevention
have not been determined. However, the risk of
alcohol-induced atrial fibrillation can be minimized by avoiding binge drinking and excessive
doses of alcohol, and by consuming <30g alcohol
daily (Table 25–17).492,493 Moretti et al.524 reported
the case of a patient treated with intermittent
courses of high doses of intravenous methylprednisolone for multiple sclerosis in whom recurrent
atrial fibrillation attributed to the corticosteroid
developed. Pretreatment prophylaxis with sotalol
was ineffective, but pretreatment prophylaxis with
oral propafenone 300 mg three times daily was
effective at preventing methylprednisoloneinduced atrial fibrillation.
MANAGEMENT
The causative agent should be discontinued. In a
series of cases of adenosine-induced atrial fibrillation, 67% of patients converted to sinus rhythm
spontaneously, whereas 33% required cardioversion.475 Therefore, hemodynamically stable druginduced atrial fibrillation or atrial flutter is
associated with a high incidence of spontaneous
conversion, and treatment may not be necessary. In
patients who do not convert spontaneously to sinus
rhythm, intravenous drugs for ventricular rate control should be administered (Table 25–18).564 For
patients with normal left ventricular function, an
intravenous b-blocker or calcium-channel blocker
should be administered. Diltiazem may be preferred
over verapamil as a result of a lower risk of symptomatic hypotension.565 For patients with heart failure due to left ventricular dysfunction, intravenous
calcium-channel blockers and b-blockers should be
avoided because of the risk of exacerbation of heart
failure; intravenous digoxin or amiodarone should
be administered instead.564 In patients with rapid
heart rates that do not respond promptly to drug
therapy for ventricular rate control, or in patients
with atrial fibrillation/flutter in whom symptoms
are unacceptable, direct-current cardioversion
should be administered. However, if atrial fibrillation has been present for >48 hours, or if the duration of the episode of atrial fibrillation is unknown,
patients should undergo transesophageal echocardiography to rule out an atrial thrombus before
electrical or pharmacologic cardioversion is performed.
After the administration of agents to control
the ventricular rate, drug-induced atrial fibrillation
or flutter should be converted to sinus rhythm,
using either elective direct-current cardioversion or
pharmacologic cardioversion. Acceptable agents
for pharmacologic cardioversion include amiodarone, dofetilide, flecainide, ibutilide, or
propafenone (Table 25–18). If the atrial fibrillation
was induced by a sodium channel blocking agent,
conversion to sinus rhythm should be achieved
using direct-current cardioversion or ibutilide. It
should be noted, however, that the effectiveness of
antiarrhythmic drug therapy for drug-induced atrial fibrillation or atrial flutter has not been studied.
In patients with atrial fibrillation in whom new
atrial flutter induced by amiodarone or
propafenone develops, radiofrequency catheter
ablation of the isthmus between the tricuspid
annulus and the inferior vena cava has been used
to terminate the atrial flutter while allowing
patients to remain on antiarrhythmic drug therapy
for the management of the atrial fibrillation.498,499
Radiofrequency catheter ablation of drug-induced
atrial flutter is successful in 90% to 100% of
patients, and drug-induced atrial flutter did not
recur in 14 of 15 patients during a 1-year follow-up
period.498
If the drug-induced atrial fibrillation is a result
of a theophylline overdose, activated charcoal may
460
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–18
Treatment Options for Drug-Induced Atrial Fibrillation564
Discontinue the offending agent.
Drug/Treatment
Recommended Doses
Ventricular rate control in patients with normal left ventricular function
Esmolol
Loading dose: 500 mcg/kg IV over 1 min
Maintenance infusion: 60–200 mcg/kg/min IV
Metoprolol
Propranolol
2.5–5 mg IV bolus over 2 min; up to 3 doses
0.15 mg/kg IV
Diltiazem
Loading dose: 0.25 mg/kg IV over 2 min
Maintenance infusion: 5–15 mg/hour IV
Verapamil
0.075–0.15 mg/kg IV over 2 min
Ventricular rate control in patients with heart failure due to left ventricular systolic dysfunction
Digoxin
0.25 mg IV every 2 hr, up to total dose of 1.5 mg
Amiodarone
Conversion to sinus rhythm
Direct-current cardioversion
Amiodaroneb
Dofetilide
Flecainideb
Ibutilide
Propafenoneb
Loading dose: 150 mg IV over 10 min
Maintenance infusion: 0.5–1 mg/min IV
200 J initially, followed by 360 J, if necessarya
Loading dose: 5–7 mg/kg IV over 30–60 min
Maintenance dose: 1.2–1.8 g daily by continuous IV infusion
CrCL >60 mL/min: 500 mcg orally twice daily
CrCL 40–60 mL/min: 250 mcg orally twice daily
CrCL 20–40 mL/min: 125 mcg orally twice daily
CrCL <20 mL/min: contraindicated
200–300 mg single oral dose
1 mg IV over 10 min. Repeat with a second 1 mg dose 10 min after the first dose if
atrial fibrillation persists.
600 mg single oral dose
CrCL = creatinine clearance; IV = intravenously.
a
Applies to both monophasic and biphasic shocks.
b
If the cause of the atrial fibrillation is a sodium channel blocking drug (amiodarone, flecainide, propafenone), avoid the use of additional sodium channel blocking agents and administer direct-current cardioversion or ibutilide. If the atrial fibrillation is caused by
theophylline toxicity: administer activated charcoal 50–100 g, followed by 50 g every 4 hr. Alternatively, continuous hemodialysis with
filtration may be performed.
be administered.566 Alternatively, continuous hemodialysis with filtration may be performed.542
INFORMATION FOR PATIENTS
Patients who are taking drugs known to cause atrial fibrillation or flutter should be instructed that
the medication, in rare instances, may cause the
heart rate (pulse) to become faster. Patients should
take their pulse daily, and should be instructed to
consult their pharmacist or physician if the heart
rate increases to above 100-120 bpm, or if they feel
palpitations, light-headed, dizzy, tired, weak, or
short of breath or experience chest pain.
ATRIAL TACHYCARDIA
Atrial tachycardia is a supraventricular arrhythmia
that is characterized on ECG by narrow QRS complexes, at a rate usually between 100 and 250
bpm.567 There are two primary types of atrial tachycardia: focal atrial tachycardia (which will be
referred to simply as “atrial tachycardia”), in which
the arrhythmia is initiated at a single atrial focus,
and multifocal atrial tachycardia, in which there
are multiple atrial foci. Focal atrial tachycardia is
characterized by regular intervals between QRS
complexes on ECG, while multifocal atrial tachycardia is associated with irregular QRS intervals and
three or more P wave morphologies.567 Atrial
CHAPTER 25 • Supraventricular Arrhythmias
TABLE 25–19
461
Agents Implicated in Drug-Induced Atrial Tachycardia
Drug
Incidence
Level of Evidence
(see page xii for explanation)
Albuterol483
Caffeine568,569
Digoxin83,388,570-580
Phenylpropanolamine569
Terbutaline581
Theophylline537,581-586
a
NK
NK
0 –4%
NK
NK
0–16%a
C
C
B
C
C
B
Incidence increases with increasing serum theophylline concentration
tachycardia may occur intermittently, which is
commonly referred to as “paroxysmal atrial tachycardia.”
CAUSATIVE AGENTS
Drugs that have been reported to cause atrial
tachycardia are listed in Table 25–19.83,388,483,537,568586
EPIDEMIOLOGY
Paroxysmal atrial tachycardia is often described as
a characteristic arrhythmia associated with digoxin
toxicity.578 However, at one large urban medical
center, “definite” paroxysmal atrial tachycardia
was documented in only 2% of 219 patients with a
discharge diagnosis of digoxin intoxication.83 In
this analysis, paroxysmal atrial tachycardia
occurred less frequently than any other arrhythmia
associated with digoxin intoxication. Therefore,
TABLE 25–20
despite the commonly held belief that paroxysmal
atrial tachycardia is commonly associated with
digoxin toxicity, it appears to occur relatively
rarely.
The incidence of multifocal atrial tachycardia
associated with theophylline is dependent on
serum theophylline concentrations.586 In an analysis of 100 patients receiving theophylline, atrial
tachycardia did not occur in the patients with
serum theophylline concentrations <10 mcg/mL.
However, in patients with serum theophylline concentrations between 10 and 20 mcg/mL, the incidence of atrial tachycardia was 8%, and the
incidence rose to 16% in patients with serum theophylline concentrations >20 mcg/mL.586
MECHANISMS
Mechanisms of drug-induced atrial tachycardia are
not entirely clear, but are likely related to increasing the automaticity of atrial tissue (Table
25–20).567 This may occur because of b-receptor
Mechanisms of Drug-Induced Atrial Tachycardia
Drug
Albuterol, terbutaline
Caffeine, theophylline
Digitalis glycoside toxicity
Mechanism
Stimulation of b-receptors, leading to enhanced atrial
automaticity
Phosphodiesterase inhibition, leading to increased plasma
concentrations of cyclic adenosine monophosphate, resulting in enhanced atrial automaticity
Extreme inhibition of the sodium–potassium–adenosine
triphosphatase pump, leading to markedly increased
myocyte concentrations of sodium, which is exchanged
with calcium, ultimately resulting in markedly increased
myocyte calcium concentrations, promoting enhanced
atrial automaticity
462
SECTION VI • Drug-Induced Cardiovascular Diseases
stimulation associated with drugs such as albuterol
or terbutaline or through phosphodiesterase inhibition and the resulting elevated concentrations of
cyclic adenosine monophosphate associated with
caffeine or theophylline.
The mechanism of paroxysmal atrial tachycardia with AV nodal blockade associated with
digitalis glycoside toxicity is likely related to
extreme inhibition of the sodium–potassium–
adenosine triphosphatase pump. This leads to
markedly increased myocyte sodium concentrations, which in turn is exchanged with calcium,
ultimately resulting in markedly increased
myocyte concentrations of calcium. This promotes enhanced atrial automaticity due to
delayed phase-4 repolarization in the atrial action
potential.587
CLINICAL PRESENTATION AND
DIFFERENTIAL DIAGNOSIS
The symptoms of drug-induced atrial tachycardia
are related to heart rate and the resultant effect on
blood pressure and cardiac output (Table 25–21).
The diagnosis of drug-induced atrial tachycardia
must be made primarily on the basis of the appearance on ECG (Figure 25–7). Conditions to consider
in the differential diagnosis of drug-induced atrial
tachycardia are presented in Table 25–22.
RISK FACTORS
Risk factors for drug-induced atrial tachycardia are
presented in Table 25–23. Paroxysmal atrial tachycardia with AV nodal blockade is extremely rare at
therapeutic serum digoxin concentrations, but
occurs primarily when serum digoxin concentrations increase to >2.0 ng/mL. Therefore, risk factors
for digoxin-induced paroxysmal atrial tachycardia
TABLE 25–21 Signs and Symptoms Associated
with Drug-Induced Atrial Tachycardia
Palpitations
Dizziness
Light-headedness
Shortness of breath
Chest pain (if underlying coronary artery disease is
present)
• Near-syncope
• Syncope
•
•
•
•
•
V1
V2
V3
FIGURE 25–7. Paroxysmal atrial tachycardia with varying
AV nodal blockade induced by digitalis toxicity. Arrows are
pointing to p waves, which are occurring at regular intervals.
Reproduced with permission from Storstein and Rasmussen.571
with AV block are the same as those for digoxin
toxicity. Since digoxin is cleared primarily by the
kidneys, renal dysfunction is a primary risk factor
for digoxin toxicity and digoxin-induced atrial
tachycardia with AV nodal blockade. In addition,
drug interactions that result in increased serum
digoxin concentrations enhance the risk of digoxin-related arrhythmias. Amiodarone,456 verapamil,457 and quinidine458 are known to
significantly increase serum digoxin concentrations.
Paroxysmal atrial tachycardia with AV block associated with digitalis toxicity may occur in patients
across a wide range of ages. In one series, in which 31
cases of paroxysmal atrial tachycardia with AV block
were reported in association with digitoxin, the age
range of patients varied from 20 to 73 years (average,
52).571 There were 15 women and 16 men, with a variety of underlying cardiovascular disorders, including
valvular heart disease (n = 14), congenital heart diseases (n = 6), and ischemic heart disease (n = 1).571 As
mentioned previously, elevated serum theophylline
concentration is a risk factor for theophylline-
TABLE 25–22 Conditions to Consider in the
Differential Diagnosis of Drug-Induced Atrial
Tachycardia
•
•
•
•
•
•
Atrial fibrillation
Atrial flutter
Atrial tachycardia not induced by drugs
Sinus tachycardia
AV nodal reentrant tachycardia
Ventricular tachycardia
CHAPTER 25 • Supraventricular Arrhythmias
463
TABLE 25–23 Risk Factors for Drug-Induced
Atrial Tachycardia
TABLE 25–24 Approaches to Help Prevent DrugInduced Atrial Tachycardia
Digoxin
• Serum digoxin concentrations >2.0 ng /mL
• Kidney disease
• Drug interactions leading to elevated serum digoxin
concentrations
Theophylline
• Serum theophylline concentrations >20 mcg /mL
Digoxin
• Maintain serum digoxin concentrations <2.0 ng /mL
• Appropriate dose adjustment for kidney disease
• Appropriate dose adjustment in the presence of interacting drugs, particularly amiodarone, verapamil, or
quinidine
Theophylline
• Maintain serum theophylline concentrations <20
mcg /mL
induced atrial tachycardia. New supraventricular
arrhythmias including multifocal atrial tachycardia
were reported in 7 patients with serum theophylline
concentrations between 21 and 40 mcg/mL.585
MORBIDITY AND MORTALITY
Patients with paroxysmal atrial tachycardia with AV
nodal blockade may present to the emergency
department and require hospitalization for management of digoxin toxicity. The incidence of mortality
associated with digitalis-induced paroxysmal atrial
fibrillation and AV nodal blockade has not been
reported, but is likely extremely low. In the series of
31 patients with digitoxin-induced paroxysmal atrial tachycardia, two patients died, but neither death
appears to have been related to the arrhythmia or
the digitoxin toxicity.571 Patients with atrial tachycardia associated with other drugs may also present
to the emergency department for treatment, particularly in the event of theophylline toxicity.
PREVENTION
Digoxin-associated paroxysmal atrial tachycardia
with AV nodal blockade is usually preventable by
maintaining serum digoxin concentrations <2.0
ng/mL (Table 25–24). Measurement of serum digoxin
concentrations is therefore recommended in patients
who may be at risk for elevated concentrations,
including patients with kidney disease or changing
kidney function and those receiving concomitant
therapy with drugs known to increase serum digoxin
concentrations (particularly amiodarone, verapamil,
or quinidine). In patients with kidney disease, appropriate dose adjustment should be made. In general,
patients with calculated creatinine clearance588 ≤20
mL/min should received an initial reduced digoxin
dose of 0.125 mg daily.589 In patients with calculated
creatinine clearance <20 mL/min who are receiving
concomitant therapy with drugs known to inhibit
the elimination of digoxin, the initial digoxin dose
should be 0.125 mg every other day.
Atrial tachycardia associated with theophylline
may be prevented by maintaining serum theophylline concentrations <20 mcg/mL.
MANAGEMENT
Treatment options for drug-induced atrial tachycardia are presented in Table 25–25. Digoxin-associated paroxysmal atrial tachycardia with AV nodal
blockade should be managed by withholding
digoxin therapy until serum digoxin concentrations decline to <2.0 ng/mL. Because paroxysmal
atrial tachycardia with AV block is rarely lifethreatening, it is usually not necessary to administer digoxin immune antibody fragments. In the
very rare event that the arrhythmia is hemodynamically unstable, synchronized direct current
cardioversion should be used to terminate the
arrhythmia. Attempts to sedate the patient should
be made, unless the urgency of the situation does
not permit them. Initial cardioversion energy
should be 50 J. Subsequent cardioversion attempts
should be performed with 100, 200, 300, and 360
J. Following the conversion of hemodynamically
unstable paroxysmal atrial tachycardia with AV
block, digoxin immune antibody fragments should
be administered to prevent recurrence of the
hemodynamically unstable arrhythmia.
Drug-induced atrial tachycardia may be
responsive to treatment with adenosine.590 In addition, drug-induced atrial tachycardia may be treated with verapamil583 or metoprolol.591 In one
study, intravenous metoprolol was more effective
than verapamil for termination of atrial tachycardia.592 Intravenous magnesium has also been effective for managing multifocal atrial tachycardia,593,594 although in one study it was not more
effective than placebo.595 If the drug-induced atrial
464
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–25
Management of Drug-Induced Atrial Tachycardia
Discontinue the offending agent.
Drug
Recommended Doses
Hemodynamically unstable
Synchronized DC cardioversion (initial energy 50 J, followed by 100 J, 200 J, 300 J,
360 J, if necessary)
If due to digoxin toxicity:
Administer digoxin immune antibody fragments. The dose is based on the serum
digoxin concentration:
Number of 38-mg vials = serum digoxin concentration x weight (kg) 4 100.
If the arrhythmia is a result of an acute digoxin overdose and the amount of digoxin
ingested is known, the dose of digoxin immune antibody fragments should be calculated as follows:
Number of 38-mg vials = mg digoxin ingested acutely x 0.8 4 0.5.
If due to theophylline toxicity:
Administer activated charcoal: 50–100 g, followed by 50 g every 4 hr
Hemodynamically stable
Metoprolol 25–50 mg single oral dose
or
5 mg IV x two doses, each administered over 5 min, doses separated by 20 min
Verapamil 5 mg IV administered over 2–5 min; second 5-mg IV dose may be administered 20 min later
Magnesium 2 g IV administered over 10 min
DC = direct current; IV = intravenously; J = Joules.
tachycardia is a result of a theophylline overdose,
activated charcoal may administered.566
ECG by regular, narrow QRS complexes, often with
an absence of discernible p waves (Figure 25–8).
INFORMATION FOR PATIENTS
CAUSATIVE AGENTS
Patients who are taking drugs that are known to
cause atrial tachycardia should be instructed that
the drug may in rare cases cause the heart rate
(pulse) to become faster. Patients should be taught
to take their pulse and to monitor their heart rate
daily. Patients should be instructed to consult their
pharmacist or physician if the heart rate increases
above 100 to 120 bpm, or if they feel light-headed,
dizzy, tired, weak, or short of breath or experience
chest pain.
Drugs that have been associated with AVNRT are
listed in Table 25–26.483,506,509,539,585,596-632
AV NODAL RECIPROCATING
TACHYCARDIA
AV nodal reciprocating tachycardia (AVNRT) is the
most common form of the group of arrhythmias
commonly referred to as “paroxysmal supraventricular tachycardia.” AVNRT is characterized on
EPIDEMIOLOGY
The incidence of drug-induced AVNRT is largely
unknown. The incidence of AVNRT associated with
albuterol and dobutamine is presented in Table
25–26.
MECHANISMS
Mechanisms of drug-induced AVNRT are somewhat unclear. Non–drug-related AVNRT was previously thought to be caused by a reentrant circuit
involving the AV node, in which there is antegrade conduction over the slow-conducting por-
CHAPTER 25 • Supraventricular Arrhythmias
FIGURE 25–8. AV nodal reciprocating tachycardia at a
rate of 200 bpm associated with fluoxetine.
465
provoke AV nodal reentry. In cases of AVNRT that
occurred in association with continuous
furosemide infusions in infants or children after
cardiac surgery, the authors speculated that a
rapid change in intravascular volume may have
contributed to the development of the arrhythmias, possibly as a result of intercompartmental
electrolyte shifts.626 Overall, the mechanisms by
which drug-induced AVNRT occurs have not been
determined, but are likely related to stimulation
of premature atrial depolarizations, alterations in
AV nodal conduction velocity or repolarization,
or both.
Reproduced with permission from Gardner et al.624
tion of the circuit and retrograde conduction over
the faster-conducting portion.633,634 However, it is
now recognized that the reentrant circuit often is
not confined to the AV node, but involves perinodal tissue.567 AV nodal or perinodal reentry is
usually initiated by a premature atrial impulse
that “uncovers” the latent reentrant circuit by
creating the circumstances required for reentry,
namely creating unidirectional block in the fast
pathway and further slowing of impulse conduction down the slow pathway. Therefore, it is possible that drugs that cause AVNRT do so by
stimulating premature atrial depolarizations that
TABLE 25–26
CLINICAL PRESENTATION AND
DIFFERENTIAL DIAGNOSIS
The symptoms of drug-induced AVNRT are similar
to those of other tachyarrhythmias (Table 25–27),
and are related to heart rate and the resultant effect
on blood pressure and cardiac output. Ventricular
rates associated with drug-induced AVNRT can be
quite fast, ranging from 150 to 300 bpm.625,626,631
Conditions to consider in the differential diagnosis
of drug-induced AVNRT are presented in Table
25–28. The diagnosis of AVNRT must be made primarily on the basis of the appearance on ECG
(Figure 25–8).
Agents Implicated in Drug-Induced Atrioventricular Nodal Reciprocating Tachycardia
Drug
Incidence
Level of Evidence
(see page xii for explanation)
Albuterol483,596-604
Caffeine605-607,b
Carbamazepine608
Clozapine609
Dobutamine506,509,610-623
Enoximone610
Fluoxetine624, 625
Furosemide626
Ipratropium627,628
Methylprednisolone629,630
631
Phenylpropanolamine
Theophylline483,539,583,585,596,632
0–21%a
NK
NK
NK
0–12%
NK
NK
NK
0.5%
NK
NK
NKa
NK = not known.
a
Incidence reported to be up to 76% in patients receiving concomitant therapy with albuterol and theophylline.
b
Suicide attempt or overdose, 2.7–35 g caffeine ingested; 1 case with intravenous caffeine administration.
A
C
C
C
A
A
C
C
A
C
C
C
466
SECTION VI • Drug-Induced Cardiovascular Diseases
TABLE 25–27 Signs and Symptoms Associated
with Drug-Induced Atrioventricular Nodal
Reciprocating Tachycardia
•
•
•
•
•
Palpitations
Dizziness
Light-headedness
Shortness of breath
Chest pain (if underlying coronary artery disease is
present)
• Near-syncope
• Syncope
TABLE 25–28 Conditions to Consider in the
Differential Diagnosis of Drug-Induced
Atrioventricular (AV) Nodal Reciprocating
Tachycardia
•
•
•
•
•
•
Atrial fibrillation
Atrial flutter
Atrial tachycardia
Sinus tachycardia
AV nodal reentrant tachycardia not induced by drugs
Ventricular tachycardia
RISK FACTORS
Known or potential risk factors for drug-induced
AVNRT are presented in Table 25–29. Albuterolinduced AVNRT appears to be more likely in
patients receiving the drug intravenously, particularly at infusion rates >60 mcg/min.597,603 AVNRT
associated with dobutamine occurs more commonly in men than in women,615 and in elderly
patients.509,616 In addition, severity of myocardial
ischemia has been shown to be an independent
predictor of dobutamine-induced AVNRT; in a
study of 1,076 patients undergoing dobutamine
stress myocardial perfusion imaging, the mean
myocardial infarction score (calculated based on
assessment of fixed perfusion defects during
myocardial perfusion scintigraphy) was 4.4±4.1 in
patients with AVNRT, as compared with 2.8±3.5 in
those without it, and the infarction score was an
independent predictor of AVNRT.616
All of the reported cases of AVNRT associated
with continuous furosemide infusions (n = 3
patients) have occurred in infants or children
after cardiac surgery. In these patients, the
arrhythmia occurred within 3 to 7 hours after the
initiation of a furosemide infusion of 1.0
TABLE 25–29 Risk Factors for Drug-Induced
Atrioventricular Nodal Reciprocating Tachycardia
Albuterol
• Continuous intravenous infusion
• Intravenous infusion rate >60 mcg/min
• Concomitant therapy with theophylline
Dobutamine
• Male sex
• Elderly
• Myocardial infarction score on dobutamine stress test
Furosemide
• Reported only in infants or children following cardiac
surgery
• Continuous infusion resulting in profound diuresis
(8–10 mL/kg /hr)
Theophylline
• Serum theophylline concentrations >20 mcg /mL
• Concomitant therapy with albuterol
mg/kg/hr. A common factor in these three
patients was substantial diuresis resulting from
the furosemide, in the range of 8 to 10 mL/kg/hr,
as compared with an average fluid elimination of
2.5 mL/kg/hr in 22 other patients who had
received a similar furosemide infusion.626 Serum
potassium concentration was below normal limits in one of the three patients in whom the
arrhythmia developed, but remained normal in
the other two cases. Therefore, it is possible that
a large amount of fluid loss may be a risk factor
or predictor of drug-induced AVNRT in infants or
children receiving a continuous infusion of
furosemide after cardiac surgery. Serum theophylline concentration is a risk factor for theophylline-induced supraventricular arrhythmias;
new supraventricular arrhythmias were reported
in 7 patients with serum theophylline concentrations between 21 and 40 mcg/mL.585 Risk factors
for AVNRT associated with other drugs have not
been identified.
MORBIDITY AND MORTALITY
Drug-induced AVNRT may result in symptoms sufficient to result in presentation to the emergency
department and hospitalization.605,606,624,628 In
addition, drug-induced AVNRT may result in prolongation of stays in the intensive care unit and
the hospital.625,626 Mortality associated with druginduced AVNRT is likely quite rare.
CHAPTER 25 • Supraventricular Arrhythmias
TABLE 25–30 Approaches to Help Prevent DrugInduced Atrioventricular Nodal Reciprocating
Tachycardia
Albuterol
• Avoid continuous intravenous infusions at rate >60
mcg/min
• Avoid concomitant therapy with theophylline
Furosemide
• Avoid diuresis of volumes >2.5 mL /kg /hr over periods
of 3–7 hr in infants/children after cardiac surgery
Theophylline
• Maintain serum theophylline concentrations <20
mcg /mL
• Avoid concomitant therapy with albuterol
PREVENTION
The likelihood of albuterol-induced AVNRT may be
reduced by avoiding continuous intravenous infusions at doses >60 mcg/min (Table 25–30).
Combinations of drugs that may induce AVNRT
should be avoided, particularly the concomitant
administration of albuterol and theophylline. In
the cases of furosemide-induced AVNRT in infants
and children after cardiac surgery, substantial fluid
elimination appears to have been a risk factor, and
in that population, avoidance of diuresis of vol-
TABLE 25–31
467
umes >2.5 mL/kg/hr over periods of 3 to 7 hours
may minimize the risk.626 Maintaining serum theophylline concentrations <20 mcg/mL reduces the
risk of theophylline-induced supraventricular
arrhythmias.
MANAGEMENT
Management options for drug-induced AVNRT are
presented in Table 25–31. As with other druginduced arrhythmias, initial management of druginduced AVNRT is discontinuation of therapy with
the offending agent. If the patient is hemodynamically unstable, synchronized direct-current cardioversion should be administered, using an initial
energy of 50 J.567 Sedation of the patient is always
preferred unless hemodynamic instability is so
severe that there is no time for sedation. If the initial conversion attempt is unsuccessful, subsequent
attempts at conversion should be made with
increasing amounts of energy (100 J, 200 J, 300 J,
360 J).567
In patients who are hemodynamically stable,
drug-induced AVNRT may respond to agents typically used to treat non–drug-induced AVNRT.
Adenosine has been found to be effective for
AVNRT associated with albuterol600,604 and may be
used as a drug of first choice for hemodynamically
Management Options for Drug-Induced AV Nodal Reciprocating Tachycardia
In hemodynamically unstable patients:
Discontinue the offending agent
Synchronized DC cardioversion (initial energy 50 J,
followed by 100 J, 200 J, 300 J, 360 J if necessary).
In hemodynamically stable patients:
Discontinue the offending agent
Vagal maneuvers (cough, carotid sinus massage, Valsalva)
Adenosine 6 mg IV rapid bolus, followed by saline flush
If no response within 2 minutes, adenosine 12 mg IV rapid
bolus, followed by saline flush
If no response within 2 minutes, adenosine 12 mg IV rapid
bolus, followed by saline flush
If no response to adenosine, and patient has LVEF > 40%
and/or no history of heart failure: Diltiazem 0.25 mg/kg IV
bolus, followed if necessary by 0.35 mg/kg IV bolus, followed by continuous infusion of 5-15 mg/hr; or verapamil
0.075-0.15 mg/kg IV over 2 minutes
If no response to adenosine, and patient has LVEF < 40%
and/or a documented history of heart failure: Digoxin 0.25
mg IV every 2 hours, up to total dose of 1.5 mg
DC = Direct current; IV = intravenously; J = Joules; LVEF = Left ventricular ejection fraction
468
SECTION VI • Drug-Induced Cardiovascular Diseases
stable AVNRT.567 Intravenous verapamil 2.5 mg has
been effective for terminating AVNRT associated
with fluoxetine.625 Intravenous verapamil should
be avoided in patients with left ventricular dysfunction567; in this population, intravenous digoxin should be administered.
INFORMATION FOR PATIENTS
Patients who are taking drugs that may cause
AVNRT should be instructed that the medication
may, in rare instances, cause the heart rate (pulse)
to become faster. Patients should be instructed to
take their pulse daily and to consult their pharmacist or physician if the heart rate increases to above
100 to 120 bpm, or if they feel palpitations, lightheaded, dizzy, tired, weak, or short of breath or
experience chest pain.
References
1. Mangrum JM, DiMarco JP. The evaluation and
management of bradycardia. N Engl J Med. 2000;342;703709.
2. Kaushik V, Leon AR, Forrester JS, et al. Bradyarrhythmias,
temporary and permanent pacing. Crit Care Med.
2000;28(Suppl):N121-N128.
3. Ufberg JW, Clark JS. Bradydysrhythmias and
atrioventricular conduction blocks. Emerg Med Clin North
Am. 2006;24:1-9.
4. DiMarco JP, Miles W, Akhtar M, et al. Adenosine for
paroxysmal supraventricular tachycardia: dose ranging
and comparison with verapamil: assessment in placebocontrolled, multicenter trials. Ann Intern Med.
1990;113:104-110.
5. Madsen CD, Pointer JE, Lynch TG. A comparison of
adenosine and verapamil for the treatment of
supraventricular tachycardia in the hospital setting. Ann
Emerg Med. 1995;25:649-655.
6. Littmann L, Anderson JD, Monroe MH. Adenosine and
Aggrenox: a hazardous combination. Ann Intern Med.
2002;137:W1.
7. Hjalmarson A, Goldstein S, Fagerberg B, et al. Effects of
controlled-release metoprolol on total mortality,
hospitalizations, and well-being in patients with heart
failure: the Metoprolol CR/XL Randomized Intervention
Trial in congestive heart failure (MERIT-HF): MERIT-HF
Study Group. JAMA. 2000;283:1295-1302.
8. Ko DT, Hebert PR, Coffey CS, et al. Adverse effects of bblocker therapy for patients with heart failure: a
quantitative overview of randomized trials. Arch Intern
Med. 2004;164:1389-1394.
9. Roy D, Talajic M, Dorian P, et al. Amiodarone to prevent
recurrence of atrial fibrillation: Canadian Trial of Atrial
Fibrillation Investigators. N Engl J Med. 2000;342:913-920.
10. The AFFIRM First Antiarrhythmic Drug Substudy
Investigators: maintenance of sinus rhythm in patients
with atrial fibrillation: an AFFIRM substudy of the first
antiarrhythmic drug. J Am Coll Cardiol. 2003;42:20-29.
11. Essebag V, Hadjis T, Platt RW, et al. Effect of amiodarone
dose on the risk of permanent pacemaker insertion.
Pacing Clin Electrophysiol. 2004;27:1519-1525.
12. Perry HE, Wright RO, Shannon MW, et al. Baclofen
overdose: drug experimentation in a group of
adolescents. Pediatrics. 1998;101:1045-1048.
13. Endmann M, Kutz R. Baclofen-intoxication in a child
resulting in coma, bradycardia, and transitory defect of
visual power. Klin Padiatr. 2005;217:89-91.
14. Leung NY, Whyte IM, Isbister GK. Baclofen overdose:
defining the spectrum of toxicity. Emerg Med Australas.
2006;18:77-82.
15. Naguib M, Magboul MM, Samarkandi AH, et al. Adverse
effects and drug interactions associated with local and
regional anaesthesia. Drug Saf. 1998;18:221-250.
16. Martinek RM. Witnessed asystole during spinal
anesthesia treated with atropine and ondansetron: a case
report. Can J Anaesth. 2004;51:226-230.
17. Takayanagi K, Hisauchi I, Watanabe J, et al.
Carbamazepine-induced sinus node dysfunction and
atrioventricular block in elderly women. Jpn Heart J.
1998;39:469-479.
18. Kaul S, Meena NK, Murthy JM. Carbamazepine induced
bradycardia. Neurol India. 2002;48:403-404.
19. Reding P, Devroede C, Barbier P. Bradycardia after
cimetidine. Lancet. 1977;2:1227.
20. Jefferys DB, Vale JA. Cimetidine and bradycardia. Lancet.
1978;1:828.
21. Hiss J, Hepler BR, Falkowski AJ, et al. Fatal bradycardia
after intentional overdose of cimetidine and diazepam.
Lancet. 1982;2:982.
22. Matthews SJ, Michelson PA, Cersosimo RJ. Cimetidineinduced sinus bradycardia. Clin Pharm. 1982;1:556558.
23. Tanner LA, Arrowsmith JB. Bradycardia and H2
antagonists. Ann Intern Med. 1988;109:434-435.
24. Cohen N, Modai D, Golik A, et al. Cimetidine-related
cardiac conduction disturbances and confusion. J Clin
Gastroenterol. 1989;11:68-69.
25. Hulisz DT, Welko JR, Heiselman DE. Sinus arrest
associated with continuous-infusion cimetidine.
Pharmacotherapy. 1993;13:64-67.
26. Schlaeffer F, Tovi F, Leiberman A. Cisplatin-induced
bradycardia. Drug Intell Clin Pharm. 1983;17:899-901.
27. Rowinsky EK, Gilbert MR, McGuire WP, et al. Sequences
of taxol and cisplatin: a phase I and pharmacologic
study. J Clin Oncol. 1991;9:1692-1703.
28. Altundağ O, Celik I, Kars A. Recurrent asymptomatic
bradycardia episodes after cisplatin infusion. Ann
Pharmacother. 2001;35:641-642.
29. Favre MP, Sztajzel J, Bertschy G. Bradycardia during
citalopram treatment: a case report. Pharmacol Res.
1999;39:149-150.
30. Rothenhäusler HB, Hoberl C, Ehrentrout S, et al. Suicide
attempt by pure citalopram overdose causing long-lasting
severe sinus bradycardia, hypotension and syncopes:
successful therapy with a temporary pacemaker.
Pharmacopsychiatry. 2000;33:150-152.
31. Isbister GK, Prior FH, Foy A. Citalopram-induced
bradycardia and presyncope. Ann Pharmacother.
2001;35:1552-1555.
32. Pai GS, Lipsitz DJ. Clonidine poisoning. Pediatrics.
1976;58:749-750.
33. MacFaul R, Miller G. Clonidine poisoning in children.
Lancet. 1977;1:1266-1267.
34. van Etta L, Burchell H. Severe bradycardia with
clonidine. JAMA. 1978;240:2047.
CHAPTER 25 • Supraventricular Arrhythmias
35. Neuvonen PJ, Vilska J, Keränen A. Severe poisoning in a
child caused by a small dose of clonidine. Clin Toxicol.
1979;14:369-374.
36. Anderson RJ, Hart GR, Crumpler CP, et al. Clonidine
overdose: report of six cases and review of the literature.
Ann Emerg Med. 1981;10:107-112.
37. Artman M, Boerth RC. Clonidine poisoning: a complex
problem. Am J Dis Child. 1983;137:171-174.
38. Olsson JM, Pruitt AW. Management of clonidine
ingestion in children. J Pediatr. 1983;103:646-650.
39. Marruecos L, Roglan A, Frati ME, et al. Crit Care Med.
1983;11:959-960.
40. Schwartz E, Friedman E, Mouallem M, et al. Sinus arrest
associated with clonidine therapy. Clin Cardiol.
1988;11:53-54.
41. Byrd BF 3rd, Collins HW, Primm RK. Risk factors for
severe bradycardia during oral clonidine therapy for
hypertension. Arch Intern Med. 1988;148:729-733.
42. Dawson PM, Vander Zanden JA, Werkman SL, et al.
Cardiac dysrhythmia with the use of clonidine in
explosive disorder. DICP. 1989;23:465-466.
43. Bonnet F, Boico O, Rostaing S, et al. Clonidine-induced
analgesia in postoperative patients: epidural versus
intramuscular administration. Anesthesiology.
1990;72:423-427.
44. Wiley JF 2nd, Wiley CC, Torrey SB, et al. Clonidine
poisoning in young children. J Pediatr. 1990;116:654-658.
45. Heidemann SM, Sarnaik AP. Clonidine poisoning in
children. Crit Care Med. 1990;18:618-620.
46. Harris JM. Clonidine patch toxicity. DICP. 1990;24:11911194.
47. Raber JH, Shinar C, Finkelstein S. Clonidine patch
ingestion in an adult. Ann Pharmacother. 1993;27:719722.
48. Rosenblum D. Clonidine-induced bradycardia in patients
with spinal cord injury. Arch Phys Med Rehabil.
1993;74:1206-1207.
49. Golusinski LL Jr, Blount BW. Clonidine-induced
bradycardia. J Fam Pract. 1995;41:399-401.
50. Owen MD, Fibuch EE, McQuillan R, et al. Postoperative
analgesia using a low-dose, oral-transdermal clonidine
combination: lack of clinical efficacy. J Clin Anesth.
1997;9:8-14.
51. Van Elstraete AC, Pastureau F, Lebrun T, et al. Caudal
clonidine for postoperative analgesia in adults. Br J
Anaesth. 2000;84:401-402.
52. Nishina K, Mikawa K, Uesugi T, et al. Efficacy of
clonidine for prevention of perioperative myocardial
ischemia: a critical appraisal and meta-analysis of the
literature. Anesthesiol. 2002;96:323-329.
53. Klein-Schwartz W. Trends and toxic effects from pediatric
clonidine exposures. Arch Pediatr Adolesc Med.
2002;156:392-396.
54. Sinha Y, Cranswick NE. Clonidine poisoning in children: a
recent experience. J Paediatr Child Health. 2004;40:678-680.
55. Spiller HA, Klein-Schwartz W, Colvin JM, et al. Toxic
clonidine ingestion in children. J Pediatr. 2005;146:263266.
56. Horowitz R, Mazor SS, Aks SE, et al. Accidental clonidine
patch ingestion in a child. Am J Ther. 2005;12:272-274.
57. Daviss WB, Patel NC, Robb AS, et al. Clonidine for
attention-deficit/ hyperactivity disorder: II. ECG changes
and adverse events analysis. J Am Acad Child Adolesc
Psychiatry. 2008;47:189-198.
58. Rangan C, Everson G, Cantrell FL. Central alpha-2
adrenergic eye drops: case series of 3 pediatric systemic
poisonings. Pediatr Emerg Care. 2008;24:167-169.
469
59. Pitner JK, Mintzer JE, Pennypacker LC, et al. Efficacy and
adverse effects of clozapine in four elderly psychotic
patients. J Clin Psychiatry. 1995;56:180-185.
60. Tupala E, Niskanen L, Tiihonen J. Transient syncope and
ECG changes associated with the concurrent
administration of clozapine and diazepam. J Clin
Psychiatry. 1999;60:619-620.
61. Castro VJ, Nacht R. Cocaine-induced bradyarrhythmia: an
unsuspected cause of syncope. Chest. 2000;117:275-277.
62. Aantaa R, Kanto J, Scheinin M. Intramuscular
dexmedetomidine, a novel alpha 2-adrenoceptor agonist,
as premedication for minor gynaecological surgery. Acta
Anaesthesiol Scand. 1991;35:283-288.
63. Aho MS, Erkola OA, Scheinin H, et al. Effect of
intravenously administered dexmedetomidine on pain
after laparoscopic tubal ligation. Anesth Analg.
1991;73:112-118.
64. Aantaa R, Jaakola ML, Kallio A, et al. A comparison of
dexmedetomidine, and alpha 2-adrenoceptor agonist,
and midazolam as i.m. premedication for minor
gynaecological surgery. Br J Anaesth. 1991;67:402-409.
65. Aho M, Scheinin M, Lehtinen AM, et al. Intramuscularly
administered dexmedetomidine attenuates
hemodynamic and stress hormone responses to
gynecologic laparoscopy. Anesth Analg. 1992;75:932-939.
66. Scheinin H, Jaakola ML, Sjövall S, et al. Intramuscular
dexmedetomidine as premedication for general
anesthesia: a comparative multicenter study.
Anesthesiology. 1993;78:1065-1075.
67. Erkola O, Korttila K, Aho M, et al. Comparison of
intramuscular dexmedetomidine and midazolam
premedication for elective abdominal hysterectomy.
Anesth Analg. 1994;79:646-653.
68. Talke P, Li J, Jain U, et al. Effects of perioperative
dexmedetomidine infusion in patients undergoing
vascular surgery: the Study of Perioperative Ischemia
Research Group. Anesthesiology. 1995;82:620-633.
69. Levänen J, Mäkelä ML, Scheinin H. Dexmedetomidine
premedication attenuates ketamine-induced
cardiostimulatory effects and postanesthetic delirium.
Anesthesiology. 1995;82:1117-1125.
70. Lawrence CJ, De Lange S. Effects of a single pre-operative
dexmedetomidine dose on isoflurane requirements and
peri-operative haemodynamic stability. Anaesthesia.
1997;52:736-744.
71. Berkenbosch JW, Tobias JD. Development of bradycardia
during sedation with dexmedetomidine in an infant
concurrently receiving digoxin. Pediatr Crit Care Med.
2003;4:203-205.
72. Dasta JF, Kane-Gill SL, Durtschi AJ. Comparing
dexmedetomidine prescribing patterns and safety in the
naturalistic setting versus published data. Ann
Pharmacother. 2004;38:1130-1135.
73. Karaaslan D, Peker TT, Alaca A, et al. Comparison of
buccal and intramuscular dexmedetomidine
premedication for arthroscopic knee surgery. J Clin
Anesth. 2006;18:589-593.
74. Coskuner I, Tekin M, Kati I, et al. Effects of
dexmedetomidine on the duration of anaesthesia and
wakefulness in bupivacaine epidural block. Eur J
Anaesthesiol. 2007;24:535-540.
75. MacLaren R, Forrest LK, Kiser TH. Adjunctive
dexmedetomidine therapy in the intensive care unit: a
retrospective assessment of impact on sedative and
analgesic requirements, levels of sedation and analgesia,
and ventilatory and hemodynamic parameters.
Pharmacotherapy. 2007;27:351-359.
470
SECTION VI • Drug-Induced Cardiovascular Diseases
76. Gómez-Vázquez ME, Hernández-Salazar E, HernándezJiménez A, et al. Clinical analgesic efficacy and side
effects of dexmedetomidine in the early postoperative
period after arthroscopic knee surgery. J Clin Anesth.
2007;19:576-582.
77. Biccard BM, Goga S, de Beurs J. Dexmedetomidine and
cardiac protection for non-cardiac surgery: a metaanalysis of randomised controlled trials. Anaesthesia.
2008;63:4-14.
78. Mason KP, Zurakowski D, Zgleszewski SE, et al. High dose
dexmedetomidine as the sole sedative for pediatric MRI.
Paediatr Anaesth. 2008;18:403-411.
79. Carroll CL, Krieger D, Campbell M, et al. Use of
dexmedetomidine for sedation of children
hospitalized in the intensive care unit. J Hosp Med.
2008;3:142-147.
80. Margolis JR, Strauss HC, Miller HC, et al. Digitalis and
the sick sinus syndrome: clinical and electrophysiologic
documentation of severe toxic effect on sinus node
function. Circulation. 1975;52:162-169.
81. Williams P, Aronson J, Sleight P. Is a slow pulse-rate a
reliable sign of digitalis toxicity? Lancet. 1978;2:13401342.
82. Rossi R, Leititis JU, Hagel KJ, et al. Severe digoxin
intoxication in a child treated by infusion of digoxinspecific Fab-antibody-fragments. Eur J Pediatr.
1984;142:138-140.
83. Mahdyoon H, Battilana G, Rosman H, et al. The evolving
pattern of digoxin intoxication: observations at a large
urban hospital from 1980 to 1988. Am Heart J.
1990;120:1189-1194.
84. Valette H, Barnay C, Lopez M, et al. Effects of
intravenous diltiazem on sinus node function and
atrioventricular conduction in patients. J Cardiovasc
Pharmacol. 1983;5:62-66.
85. Schroeder JS, Hung J, Lamb IH, et al. Diltiazem and
propranolol, alone and in combination, on exercise
performance and left ventricular function in patients
with stable effort angina: a double-blind, randomized,
and placebo-controlled study. Acta Pharmacol Toxicol.
1985;57(Suppl 2):55-60.
86. Gibson RS, Boden WE, Theroux P, et al. Diltiazem and
reinfarction in patients with non-Q-wave myocardial
infarction: results of a double-blind, randomized,
multicenter trial. N Engl J Med. 1986;315:423-429.
87. Sagie A, Strasberg B, Kusnieck J, et al. Symptomatic
bradycardia induced by the combination of oral
diltiazem and beta blockers. Clin Cardiol. 1991;14:314316.
88. Felicetta JV, Serfer HM, Cutler NR, et al. A dose-response
trial of once-daily diltiazem. Am Heart J. 1992;123:10221026.
89. Andrivet P, Beaslay V, Kiger JP, et al. Complete sinus
arrest during diltiazem therapy: clinical correlates and
efficacy of intravenous calcium. Eur Heart J. 1994;15:350354.
90. Boudonas G, Lefkos N, Efthymiadis AP, et al. Intravenous
administration of diltiazem in the treatment of
supraventricular tachyarrhythmias. Acta Cardiol.
1995;50:125-134.
91. Théroux P, Grégoire J, Chin C, et al. Intravenous
diltiazem in acute myocardial infarction: Diltiazem as
Adjunctive Therapy to Activase (DATA) trial. J Am Coll
Cardiol. 1998;32:620-628.
92. Edoute Y, Nagachandran P, Svirski B, et al. Cardiovascular
adverse drug reaction associated with combined beta-
93.
94.
95.
96.
97.
98.
99.
100.
101.
102.
103.
104.
105.
106.
107.
108.
109.
110.
111.
112.
adrenergic and calcium entry-blocking agents. J
Cardiovasc Pharmacol. 2000;35:556-559.
Shapira OM, Alkon JD, Macron DS, et al. Nitroglycerin is
preferable to diltiazem for prevention of coronary bypass
conduit spasm. Ann Thorac Surg. 2000;70:883-888.
Amorim S, Dias P, Rocha G, et al. Poisoning with calcium
channel blockers: a case report and review of the
literature. Rev Port Cardiol. 2001;20:1249-1257.
Punukollu G, Gowda RM, Khan IA, et al. Delayed
presentation of calcium channel antagonist overdose. Am
J Ther. 2003;10:132-134.
Mills TA, Kawji MM, Cataldo VD, et al. Profound sinus
bradycardia due to diltiazem, verapamil, and/or betaadrenergic blocking drugs. J La State Med Soc.
2004;156:327-331.
Pennell DJ, Underwood SR, Ell PJ. Symptomatic
bradycardia complicating the use of intravenous
dipyridamole for thallium-201 myocardial perfusion
imaging. Int J Cardiol. 1992;27:272-274.
Picano E, Marini C, Pirelli S, et al. Safety of intravenous
high-dose dipyridamole echocardiography. Am J Cardiol.
1992;15:252-258.
Bjoernstad K, Aakhus S, Lundbom J, et al. Digital
dipyridamole stress echocardiography in silent ischemia
after coronary artery bypass grafting and/or after healing
of acute myocardial infarction. Am J Cardiol.
1993;72:640-646.
Roach PJ, Magee MA, Freedman SB. Asystole and
bradycardia during dipyridamole stress testing in patients
receiving beta blockers. Int J Cardiol. 1993;42:92-94.
Lo Mauro R, Sabella FP, Enia F. Sinus arrest associated
with dipyridamole infusion. Chest. 1994;105:604-605.
Bjørnstad K, Aakhus S, Hatle L. Digital high frame rate
stress echocardiography for detection of coronary artery
stenosis by high dose dipyridamole stress testing. Int J
Card Imaging. 1995;11:163-170.
Roman MR, Angelides S, Freeman AP, et al. The safety of
dipyridamole in patients undergoing myocardial
perfusion scintigraphy prior to lung volume reduction
surgery. Eur J Nucl Med. 2001;28:1405-1408.
Hayler AM, Holt DW, O’Keeffe B, et al. Treatment of
disopyramide overdosage. Med J Aust. 1979;1:234.
Camm J, Ward D, Spurrell RA. The effect of intravenous
disopyramide phosphate on recurrent paroxysmal
tachycardias. Br J Clin Pharmacol. 1979;8:441-449.
Warrington SJ, Hamer J. Some cardiovascular problems
with disopyramide. Postgrad Med J. 1980;56:229-233.
Desai JM, Scheinman MM, Hirschfeld D, et al.
Cardiovascular collapse associated with disopyramide
therapy. Chest. 1981;79:545-551.
Shepherd G, Klein-Schwartz W, Edwards R. Donepezil
overdose: a tenfold dosing error. Ann Pharmacother.
1999;33:812-815.
Relkin NR, Reichman WE, Orazem J, et al. A large,
community-based, open-label trial of donepezil in the
treatment of Alzeheimer’s disease. Dement Geriatr Cogn
Disord. 2002;16:15-24.
Bordier P, Garrigue S, Barold SS, et al. Significance of
syncope in patients with Alzheimer’s disease treated with
cholinesterase inhibitors. Europace. 2003;5:429-431.
Bordier P, Lanusse S, Garrigue S, et al. Causes of syncope
in patients with Alzheimer’s disease treated with
donepezil. Drugs Aging. 2005;22:687-694.
Singh BN, Connolly SJ, Crijns HJ, et al. Dronedarone for
maintenance of sinus rhythm in atrial fibrillation or
flutter. N Engl J Med. 2007;357:987-999.
CHAPTER 25 • Supraventricular Arrhythmias
113. Hohnloser SH, Crijns HJ, van Eickels M, et al. Effect of
dronedarone on cardiovascular events in atrial
fibrillation. N Engl J Med. 2009;360:668-678.
114. Beyenburg S, Schönegger K. Severe bradycardia in a
stroke patient caused by a single low dose of
escitalopram. Eur Neurol. 2007;57:50-51.
115. Reid PR, Griffith LS, Platia EV, et al. Evaluation of
flecainide acetate in the management of patients at high
risk of sudden cardiac death. Am J Cardiol. 1984;53:108B111B.
116. Platia EV, Estes M, Heine DL, et al. Flecainide:
electrophysiologic and antiarrhythmic properties in
refractory ventricular tachycardia. Am J Cardiol.
1985;55:956-962.
117. Crozier IG, Ikram H, Kenealy M, et al. Flecainide acetate
for conversion of acute supraventricular tachycardia to
sinus rhythm. Am J Cardiol. 1987;59:607-609.
118. Van Gelder IC, Crijns HJGM, Van Glist WH, et al.
Efficacy and safety of flecainide acetate in the
maintenance of sinus rhythm after electrical
cardioversion of chronic atrial fibrillation or atrial flutter.
Am J Cardiol. 1989;64:1317-1321.
119. Köppel C, Oberdisse U, Heinemeyer G. Clinical course
and outcome in class IC antiarrhythmic overdose. J
Toxicol Clin Toxicol. 1990;28:433-444.
120. Hanley NA, Bourke JP, Gascoigne AD. Survival in a case
of life-threatening flecainide overdose. Intensive Care Med.
1998;24:740-742.
121. Aliot E, De Roy L, Capucci A, et al. Safety of a controlledrelease flecainide acetate formulation in the prevention
of paroxysmal atrial fibrillation in outpatients. Ann
Cardiol Angeiol. 2003;52:34-40.
122. Ellison JM, Milofsky JE, Ely E. Fluoxetine-induced
bradycardia and syncope in two patients. J Clin
Psychiatry. 1990;51:385-386.
123. Buff DD, Brenner R, Kirtane SS, et al. Dysrhythmia
associated with fluoxetine treatment in an elderly patient
with cardiac disease. J Clin Psychiatry. 1991;52:174-176.
124. Ahmed I, Dagincourt PG, Miller LG, et al. Possible
interaction between fluoxetine and pimozide causing
sinus bradycardia. Can J Psychiatry. 1993;38:62-63.
125. Hussein S, Kaufman BM. Bradycardia associated with
fluoxetine in an elderly patient with sick sinus
syndrome. Postgrad Med J. 1994;70:56.
126. Anderson J, Compton SA. Fluoxetine induced
bradycardia in presenile dementia. Ulster Med J.
1997;66:144-145.
127. Spigset O. Adverse reactions of selective serotonin
reuptake inhibitors: reports from a spontaneous
reporting system. Drug Saf. 1999;20:277-287.
128. Compton R, Spiller HA, Bosse GM. Fatal fluoxetine
ingestion with postmortem blood concentrations. Clin
Toxicol. 2005;43:277-279.
129. Chin RL, Sporer KA, Cullison B, et al. Clinical course of
gamma-hydroxybutyrate overdose. Ann Emerg Med.
1998;31:716-722.
130. Anonymous. Adverse events associated with ingestion of
gamma-butyrolactone: Minnesota, New Mexico, and
Texas, 1998-1999. MMWR Morb Mortal Wkly Rep.
1999;48:137-140.
131. Liechti ME, Kunz I, Greminger P, et al. Clinical features
of gamma-hydroxybutyrate and gamma-butyrolactone
toxicity and concomitant drug and alcohol use. Drug
Alcohol Depend. 2006;81:323-326.
132. Zuck D, Rao JJ. Profound bradycardia with verapamil and
halothane. Anaesthesia. 1985;40;84-85.
471
133. Kataria B, Epstein R, Bailey A, et al. A comparison of
sevoflurane to halothane in paediatric surgical patients:
results of a multicentre international study. Paediatr
Anaesth. 1996;6:283-292.
134. Annila P, Rorarius M, Reinikainen P, et al. Effect of pretreatment with intravenous atropine or glycopyrrolate
on cardiac arrhythmias during halothane anaesthesia for
adenoidectomy in children. Br J Anaesth. 1998;80:756760.
135. Annila P, Viitanen H, Reinikainen P, et al. Induction
characteristics of thiopentone/suxamethonium,
propofol/alfentanil or halothane alone in children aged
1-3 years. Eur J Anaesthesiol. 1999;16:359-366.
136. Amin N, Borzak S, Housholder SA, et al. Multiple
episodes of sinus arrest following the administration of
ibutilide in a patient with underlying sinus node
dysfunction. Heart. 1998;79:628-629.
137. Taylor CT, Donaldson AR. Safety and effectiveness of
ibutilide in a community hospital. Pharmacotherapy.
2001;21:488-492.
138. Gowda RM, Khan IA, Punukollu G, et al. Use of ibutilide
for cardioversion of recent-onset atrial fibrillation and
flutter in elderly. Am J Ther. 2004;11:95-97.
139. Viktorsdottir O, Henriksdottir A, Arnar DO. Ibutilide for
treatment of atrial fibrillation in the emergency
department. Emerg Med J. 2006;23:133-134.
140. Romano MJ, Gaylor A, Sang CJ Jr. Life-threatening
isradipine poisoning in a child. Pharmacotherapy.
2002;22:766-770.
141. Nettles DC, Herrin TJ, Mullen JG. Ketamine induction in
poor-risk patients. Anesth Analg. 1973;52:59-64.
142. Wolfson B, Kielar CM, Shenoy NR, et al. Analgesic
“cocktails” for pneumoencephalography: ketamine,
diazepam, alphaprodine, and droperidol. Anesth Analg.
1974;52:779-783.
143. Levin RM, Seleny FL, Streczyn MV. Ketaminepancuronium-narcotic technic for cardiovascular surgery
in infants: a comparative study. Anesth Analg.
1975;54:800-805.
144. Radnay PA, Hollinger I, Santi A, et al. Ketamine for
pediatric cardiac anesthesia. Anaesthesist. 1976;25:259-265.
145. Green SM, Denmark TK, Cline J, et al. Ketamine sedation
for pediatric critical care procedures. Pediatr Emerg Care.
2001;17:244-248.
146. Cheng TO, Wadhwa K. Sinus standstill following
intravenous lidocaine administration. JAMA.
1973;223:790-792.
147. Parameswaran R, Kahn D, Monheit R, et al. Sinus
bradycardia due to lidocaine: clinical-electrophysiologic
correlations. J Electrocardiol. 1974;7:75-78.
148. Marriott HJ, Phillips K. Profound hypotension and
bradycardia after a single bolus of lidocaine. J
Electrocardiol. 1974;7:79-82.
149. Klein HO, Jutrin I, Kaplinsky E. Cerebral and cardiac
toxicity of a small dose of lignocaine. Br Heart J.
1975;37:775-778.
150. Zoll WR, Täuber M, Vorburger C. Sinus arrest after
intravenous administration of lidocaine. Schweiz Med
Wochenschr. 1979;109:1946-1948.
151. Demczuk RJ. Significant sinus bradycardia following
intravenous lidocaine injection. Anesthesiology.
1984;60:69-70.
152. Karir V. Bradycardia associated with intravenous
methadone administered for sedation in a patient with
acute respiratory distress syndrome. Pharmacotherapy.
2002;22:1196-1199.
472
SECTION VI • Drug-Induced Cardiovascular Diseases
153. Ashwath ML, Ajjan M, Culclasure T. Methadone-induced
bradycardia. J Emerg Med. 2005:29:73-75.
154. Wheeler AD, Tobias JD. Bradycardia during methadone
therapy in an infant. Pediatr Crit Care Med. 2006;7:83-85.
155. Tvede N, Nielsen LP, Andersen V. Bradycardia after highdose intravenous methylprednisolone therapy. Scand J
Rheumatol. 1986;15:302-304.
156. Lucas KG, Howrie DL, Phebus CK. Cardiorespiratory
decompensation following methylprednisolone
administration. Pediatr Hematol Oncol. 1993;10:249-255.
157. Küçükosmanoğlu O, Karabay A, Ozbarlas N, et al. Marked
bradycardia due to pulsed and oral methylprednisolone
therapy in a patient with rapidly progressive
glomerulonephritis. Nephron. 1998;80:484.
158. Guillén EL, Ruíz AM, Bugallo JB. Hypotension,
bradycardia, and asystole after high-dose intravenous
methylprednisolone in a monitored patient. Am J Kidney
Dis. 1998;32:E4.
159. Thornton PD, Hamblin M, Treleaven JG, et al. High dose
methyl prednisolone in refractory chronic lymphocytic
leukaemia. Leuk Lymphoma. 1999;34:167-170.
160. Pudil R, Hrncir Z. Severe bradycardia after a
methylprednisolone “minipulse” treatment. Arch Intern
Med. 2001;161:1778-1779.
161. Akikusa JD, Feldman BM, Gross GJ, et al. Sinus
bradycardia after intravenous pulse methylprednisolone.
Pediatrics. 2007;119:e778-e782.
162. Feneck RO, Sherry KM, Withington PS, et al. Comparison
of the hemodynamic effects of milrinone with
dobutamine in patients after cardiac surgery. J Cardiothorc
Vasc Anesth. 2001;15:306-315.
163. Sprague DH. Severe bradycardia after neostigmine in a
patient taking propranolol to control paroxysmal atrial
tachycardia. Anesthesiology. 1975;42:208-210.
164. Seidl DC, Martin DE. Prolonged bradycardia after
neostigmine administration in a patient taking nadolol.
Anesth Analg. 1984;63:365-367.
165. Triantafillou AN, Tsueda K, Berg J, et al. Refractory
bradycardia after reversal of muscle relaxant in a diabetic
with vagal neuropathy. Anesth Analg. 1986;65:1237-1241.
166. Arsura EL, Brunner NG, Namba T, et al. Adverse
cardiovascular effects of anticholinesterase medications.
Am J Med Sci. 1987;293:18-23.
167. Eldor J, Hoffman B, Davidson JT. Prolonged bradycardia
and hypotension after neostigmine administration in a
patient receiving atenolol. Anaesthesia. 1987;42:12941297.
168. Shankar KB, Moseley H, Kumar A. Prolonged
bradycardia and hypotension after neostigmine
administration in a patient receiving atenolol.
Anaesthesia. 1988;43:797-798.
169. Backman SB, Ralley FE, Fox GS. Neostigmine produces
bradycardia in a heart transplant patient. Anesthesiology.
1993;78:777-779.
170. Backman SB, Fox GS, Stein RD, et al. Neostigmine
decreases heart rate in heart transplant patients. Can J
Anaesth. 1996;43:373-378.
171. Backman SB, Stein RD, Ralley FE, et al. Neostigmineinduced bradycardia following recent vs remote cardiac
transplantation in the same patient. Can J Anaesth.
1996;43:394-398.
172. Ponec RJ, Saunders MD, Kimmey MB. Neostigmine for
the treatment of acute colonic pseudo-obstruction. N
Engl J Med. 1999;341:137-141.
173. Abeyta BJ, Albrecht RM, Schermer CR. Retrospective
study of neostigmine for the treatment of acute colonic
pseudo-obstruction. Am Surg. 2001;67:265-268.
174. Ho KM, Ismail H, Lee KC, et al. Use of intrathecal
neostigmine as an adjunct to other spinal medications in
perioperative and peripartum analgesia: a meta-analysis.
Anaesth Intensive Care. 2005;33:41-53.
175. Kross RA, Ferri E, Leung D, et al. A comparative study
between a calcium channel blocker (Nicardipine) and a
combined alpha-beta-blocker (Labetalol) for the control
of emergence hypertension during craniotomy for tumor
surgery. Anesth Analg. 2000;91:904-909.
176. Arima H, Sobue K, Tanaka S, et al. Profound sinus
bradycardia after intravenous nicardipine. Anesth Analg.
2002;95:53-55.
177. Come PC, Pitt B. Nitroglycerin-induced severe
hypotension and bradycardia in patients with acute
myocardial infarction. Circulation. 1976;54:624-628.
178. Ong EA, Bass S. Nitroglycerin-induced bradycardia and
hypotension in acute myocardial infarction. Chest.
1980;77:244.
179. Meretoja OA. Haemodynamic effects of combined
nitroglycerin- and dobutamine-infusions after coronary
by-pass surgery: with one nitroglycerin-related
complication. Acta Anaesthesiol Scand. 1980;24:211-215.
180. Nemerovski M, Shah PK. Syndrome of severe bradycardia
and hypotension following sublingual nitroglycerin
administration. Cardiology. 1981;67:180-189.
181. Khan AH, Carleton RA. Nitroglycerin-induced
hypotension and bradycardia. Arch Intern Med.
1981;141:984.
182. Brandes W, Santiago T, Limacher M. Nitroglycerininduced hypotension, bradycardia, and asystole: report
of a case and review of the literature. Clin Cardiol.
1990;13:741-744.
183. Buckley R, Roberts R. Symptomatic bradycardia following
the administration of sublingual nitroglycerin. Am J
Emerg Med. 1993;11:253-255.
184. Wuerz R, Swope G, Meador S, et al. Safety of prehospital
nitroglycerin. Ann Emerg Med. 1994;23:31-36.
185. Engelberg S, Singer AJ, Moldashel J, et al. Effects of
prehospital nitroglycerin on hemodynamics and chest
pain intensity. Prehosp Emerg Care. 2000;4:290-293.
186. Markowitz JS, DeVane CL, Boulton DW, et al.
Hypotension and bradycardia in a healthy volunteer
following a single 5 mg dose of olanzapine. J Clin
Pharmacol. 2002;42:104-106.
187. Lee TW, Tsai SJ, Hwang JP. Severe cardiovascular side
effects of olanzapine in an elderly patient: case report. Int
J Psychiatry Med. 2003;33:399-401.
188. Chen CC, Tsai JH, Yang P, et al. Bradyarrhythmic shock
associated with olanzapine. Aust N Z J Psychiatry.
2007;41:89.
189. Escobar R, Pérez V, San L, et al. Effectiveness results of
olanzapine in acute psychotic patients with agitation in
the emergency room setting: results from NATURA study.
Actas Esp Psiquiatr. 2008;36:151-157.
190. McGuire WP, Rowinsky EK, Rosenshein NB, et al. Taxol: a
unique antineoplastic agent with significant activity in
advanced ovarian epithelial neoplasms. Ann Intern Med.
1989;111:273-279.
191. Rowinsky EK, McGuire WP, Guarnieri T, et al. Cardiac
disturbances during the administration of taxol. J Clin
Oncol. 1991;9:1704-1712.
192. Arbuck SG, Strauss H, Rowinsky E, et al. A reassessment
of cardiac toxicity associated with Taxol. J Natl Cancer
Inst Monogr. 1993;15:117-130.
193. Chamberlain MC, Kormanik P. Salvage chemotherapy
with paclitaxel for recurrent primary brain tumors. J Clin
Oncol. 1995;13:2066-2071.
CHAPTER 25 • Supraventricular Arrhythmias
194. Glück S, Germond C, Lopez P, et al. A phase I trial of
high-dose paclitaxel, cyclophosphamide and
mitoxantrone with autologous blood stem cell support
for the treatment of metastatic breast cancer. Eur J
Cancer. 1998;34:1008-1014.
195. Kanat O, Evrensel T, Baran I, et al. Protective effect of
amifostine against toxicity of paclitaxel and carboplatin
in non-small cell lung cancer: a single center randomized
study. Med Oncol. 2003;20:237-245.
196. Kamineni P, Prakasa K, Hasan SP, et al. Cardiotoxicities of
paclitaxel in African Americans. J Natl Med Assoc.
2003;95:977-981.
197. Ruiz-Casado A, Calzas J, García J, et al. Life-threatening
adverse drug reaction to paclitaxel: postmarketing
surveillance. Clin Transl Oncol. 2006;8:60-61.
198. Kietpeerakool C, Tiyayon J, Suprasert P, et al. Benefit of
electrocardiography during front-line combination
paclitaxel and carboplatin chemotherapy for epithelial
ovarian cancer. J Med Assoc Thai. 2006;89:1805-1810.
199. Rita L, Cox JM, Seleny FL, et al. Ketamine hydrochloride
for pediatric premedication: I. Comparison with
pentazocine. Anesth Analg. 1974;53:375-379.
200. Clémenty J, Coste P, Metzinger M. Efficacy and tolerance
of propafenone in the treatment of cardiac rhythm
disorders: evaluation of a multicenter open trial on 3,687
patients. Ann Cardiol Angeiol. 1987;36:207-212.
201. Lee PK, Kerr CR, Vorderbrugge S, et al. Symptomatic
sinus node dysfunction associated with the use of
propafenone. Am J Cardiol. 1988;62:480-481.
202. Kerns W 2nd, English B, Ford M. Propafenone overdose.
Ann Emerg Med. 1994;24:98-103.
203. Mörike K, Magadum S, Mettang T, et al. Propafenone in a
usual dose produces severe side-effects: the impact of
genetically determined metabolic status on drug therapy.
J Intern Med. 1995;238:469-472.
204. Thomson SJ, Yate PM. Bradycardia after propofol
infusion. Anaesthesia. 1987;42:430.
205. Baraka A. Severe bradycardia following propofolsuxamethonium sequence. Br J Anaesth. 1988;61:482-483.
206. Saarnivaara L, Hiller A, Oikkonen M. QT interval, heart
rate and arterial pressures using propofol, thiopentone or
methohexitone for induction of anaesthesia in children.
Acta Anaesthesiol Scand. 1993;37:419-423.
207. Hug CC Jr, McLeskey CH, Nahrwold ML, et al.
Hemodynamic effects of propofol: data from over 25,000
patients. Anesth Analg. 1993;77(4 Suppl):S21-S29.
208. McConaghy P, Bunting HE. Assessment of intubating
conditions in children after induction with propofol and
varying doses of alfentanil. Br J Anaesth. 1994;73:596599.
209. Tramèr MR, Moore RA, McQuay HJ. Propofol and
bradycardia: causation, frequency and severity. Br J
Anaesth. 1997;78:642-651.
210. Bassett KE, Anderson JL, Pribble CG, et al. Propofol for
procedural sedation in children in the emergency
department. Ann Emerg Med. 2003;42:773-782.
211. Ulmer BJ, Hansen JJ, Overley CA, et al. Propofol versus
midazolam/fentanyl for outpatient colonoscopy:
administration by nurses supervised by endoscopists.
Clin Gastroenterol Hepatol. 2003;1:425-432.
212. Taha S, Siddik-Sayyid S, Alameddine M, et al. Propofol is
superior to thiopental for intubation without muscle
relaxants. Can J Anaesth. 2005;52:249-253.
213. Wilson SE, Boyack DA, Ezeife-Ugorji OC, et al. Propofolinduced bradycardia in a pediatric patient undergoing
chemotherapy with the known cardiotoxic agent
cytarabine. Paediatr Anaesth. 2006;16:702-704.
473
214. Yorozu T, Iijima T, Matsumoto M, et al. Factors
influencing intraoperative bradycardia in adult patients. J
Anesth. 2007;21:136-141.
215. Sieg A. Propofol sedation in outpatient colonoscopy by
trained practice nurses supervised by the
gastroenterologist: a prospective evaluation of over 3000
cases. Z Gastroenterol. 2007;45:697-701.
216. Poon CM, Leung TL, Wong CW, et al. Safety of nurseadministered propofol sedation using PCA pump for
outpatient colonoscopy in Chinese patients: a pilot
study. Asian J Surg. 2007;30:239-243.
217. Baraka A, Dajani A. Severe bradycardia following
physostigmine in the presence of beta-adrenergic
blockade: a case report. Middle East J Anesthesiol.
1984;7:291-293.
218. Zvosec DL, Smith SW, Litonjua R, et al. Physostigmine
for gamma-hydroxybutyrate coma: inefficacy, adverse
events, and review. Clin Toxicol. 2007;45:261-265.
219. Camarri E, Chirone E, Fanteria G, et al. Ranitidineinduced bradycardia. Lancet. 1984;2:160.
220. Jack D, Richards DA, Granata F. Side-effects of ranitidine.
Lancet. 1982;2(8292):264-265.
221. Shah RR. Symptomatic bradycardia in association with
H2-receptor antagonists. Lancet. 1982;2:1108.
222. Schifman C. Gynecomastia and bradycardia: side effects
of ranitidine? Clin Pharm. 1983;2:209.
223. Balestrazzi P, Gregori G, Bernasconi S, et al. Bradycardia
and neurologic disorders associated with ranitidine in a
child. Am J Dis Child. 1985;139:442.
224. Johnson WS, Miller DR. Ranitidine and bradycardia. Ann
Intern Med. 1988;108:493.
225. Khera DC, Smith SL, Slabic SF. Ranitidine-induced
bradycardia. Am J Gastroenterol. 1988;83:332-333.
226. Nahum E, Reish O, Naor N, et al. Ranitidine-induced
bradycardia in a neonate: a first report. Eur J Pediatr.
1993;152:933-934.
227. Yang J, Russell DA, Bourdeau JE. Case report: ranitidineinduced bradycardia in a patient with dextrocardia. Am J
Med Sci. 1996;312:133-135.
228. Schüttler J, Albrecht S, Breivik H, et al. A comparison of
remifentanil and alfentanil in patients undergoing major
abdominal surgery. Anaesthesia. 1997;52:307-317.
229. DeSouza G, Lewis MC, TerRiet MF. Severe bradycardia
after remifentanil. Anesthesiology. 1997;87:1019-1020.
230. Reid JE, Mirakhur RK. Bradycardia after administration of
remifentanil. Br J Anaesth. 2000;84:422-423.
231. Elliott P, O’Hare R, Bill KM, et al. Severe cardiovascular
depression with remifentanil. Anesth Analg. 2000;91:5861.
232. Prys-Roberts C, Lerman J, Murat I, et al. Comparison of
remifentanil versus regional anaesthesia in children
anaesthetised with isoflurane/nitrous oxide:
International Remifentanil Paediatric Anaesthesia Study
group. Anaesthesia. 2000;55:870-876.
233. Chung F, Mulier JP, Scholz J, et al. A comparison of
anaesthesia using remifentanil combined with either
isoflurane, enflurane or propofol in patients undergoing
gynaecological laparoscopy, varicose vein or
arthroscopic surgery. Acta Anaesthesiol Scand.
2000;44:790-798.
234. Chinachoti T, Werawatganon T, Suksompong S, et al. A
multicenter randomized double-blind comparison of
remifentanil and alfentanil during total intravenous
anaesthesia for out-patient laparoscopic gynaecological
procedures. J Med Assoc Thai. 2000;83:1324-1332.
235. Krenn H, Deusch E, Jellinek H, et al. Remifentanil or
propofol for sedation during carotid endarterectomy
474
236.
237.
238.
239.
240.
241.
242.
243.
244.
245.
246.
247.
248.
249.
250.
251.
252.
253.
254.
SECTION VI • Drug-Induced Cardiovascular Diseases
under cervical plexus block. Br J Anaesth. 2002;89:637640.
Nilsson LB, Viby-Mogensen J, Møller J, et al.
Remifentanil vs. alfentanil for direct laryngoscopy: a
randomized study comparing two total intravenous
anaesthesia techniques: TIVA for direct laryngoscopy.
Acta Anaesthesiol Belg. 2002;53:213-219.
Breslin DS, Farling PA, Mirakhur RK. The use of
remifentanil in the anaesthetic management of patients
undergoing adrenalectomy: a report of three cases.
Anaesthesia. 2003;58:358-362.
Weale NK, Rogers CA, Cooper R, et al. Effect of
remifentanil infusion rate on stress response to the prebypass phase of paediatric cardiac surgery. Br J Anaesth.
2004;92:187-194.
Leidinger W, Schwinn P, Hofmann HM, et al.
Remifentanil for analgesia during retrobulbar nerve block
placement. Eur J Anaesthesiol. 2005:22:40-43.
Briassoulis G, Spanaki AM, Vassilaki E, et al. Potentially
life-threatening bradycardia after remifentanil infusion
in a child. Acta Anaesthesiol Scand. 2007;51:1130.
Goyal RS, Goyal SB. Symptomatic bradyarrhythmia
secondary to risperidone. Am J Psychiatry. 2003;160:2243.
Tran KT, Golden P, Lark T, et al. Bradycardia at low doses
of risperidone. Am J Psychiatry. 2004;161:2325-2326.
Raeder J, Gupta A, Pedersen FM. Recovery characteristics
of sevoflurane- or propofol-based anaesthesia for day-care
surgery. Acta Anaesthesiol Scand. 1997;41:988-994.
Townsend P, Stokes MA. Bradycardia during rapid
inhalation induction with sevoflurane in children. Br J
Anaesth. 1998;80:410.
Gravel NR, Searle NR, Taillefer J, et al. Comparison of the
hemodynamic effects of sevoflurane anesthesia induction
and maintenance vs TIVA in CABG surgery. Can J
Anaesth. 1999;46:240-246.
Green DH, Townsend P, Bagshaw O, et al. Nodal rhythm
and bradycardia during inhalation induction with
sevoflurane in infants: a comparison of incremental and
high-concentration techniques. Br J Anaesth.
2000;85:369-370.
Roodman S, Bothwell M, Tobias JD. Bradycardia with
sevoflurane induction in patients with trisomy 21.
Paediatr Anaesth. 2003;13:538-540.
Casati A, Aldegheri G, Vinciguerra E, et al. Randomized
comparison between sevoflurane anaesthesia and
unilateral spinal anaesthesia in elderly patients
undergoing orthopaedic surgery. Eur J Anaesthesiol.
2003;20:640-646.
Hodgson RE, Dawson P, Hold AR, et al. Anaesthesia for
electroconvulsive therapy: a comparison of sevoflurane
with propofol. Anaesth Intensive Care. 2004;32:241-245.
Nagata S, Kazekawa K, Aikawa H, et al. Hemodynamic
stability under general anesthesia in carotid artery
stenting. Radiat Med. 2005;23:427-431.
Taylor MH, Wilton NC. Bradycardia with sevoflurane in
siblings with Branchio-oto-renal syndrome. Paediatr
Anaesth. 2007;17:80-83.
Goldfinger MM, Tripi PA. Cardiac arrest in a child with
cerebral palsy undergoing sevoflurane induction of
anesthesia after preoperative clonidine. Paediatr Anaesth.
2007;17:270-272.
Latour Y, Dumont G, Brosseau A, et al. Effects of sotalol
in twenty patients with cardiac arrhythmias. Int J Clin
Pharmacol Biopharm. 1977;15:275-278.
Neuvonen PJ, Elonen E, Tarssanen L. Sotalol
intoxication, two patients with concentration-effect
relationships. Acta Pharmacol Toxicol. 1979;45:52-57.
255. Gupta K. Hazards of B-blockers in the elderly: severe
bradycardia due to Sotacor overdose. Br J Clin Pract.
1985;39:116-117.
256. Campbell TJ, Gavaghan TP, Morgan JJ. Intravenous
sotalol for the treatment of atrial fibrillation and flutter
after cardiopulmonary bypass: comparison with
disopyramide and digoxin in a randomised trial. Br Heart
J. 1985;54:86-90.
257. Edvardsson N, Varnauskas E. Clinical course, serum
concentrations and elimination rate in a case of massive
sotalol intoxication. Eur Heart J. 1987;8:544-548.
258. Anastasiou-Nana MI, Anderson JL, Askins JC, et al.
Long-term experience with sotalol in the treatment of
complex ventricular arrhythmias. Am Heart J.
1987;114:288-296.
259. Soyka LF, Wirtz C, Spangenberg RB. Clinical safety profile
of sotalol in patients with arrhythmias. Am J Cardiol.
1990;65:74A-81A.
260. Obel IW, Jardine R, Haitus B, et al. Efficacy of oral sotalol
in reentrant ventricular tachycardia. Cardiovasc Drugs
Ther. 1990;4(Suppl 3):613-618.
261. Tipple M, Sandor G. Efficacy and safety of oral sotalol in
early infancy. Pacing Clin Electrophysiol. 1991;14:2062-2065.
262. Horrigan MC, Davis MJ, May C, et al. Dual chamber rate
responsive pacing to allow sotalol therapy for ventricular
tachycardia. Pacing Clin Electrophysiol. 1992;15:21082110.
263. Winters SL, Kukin M, Pe E, et al. Effect of oral sotalol on
systemic hemodynamics and programmed electrical
stimulation in patients with ventricular arrhythmias and
structural heart disease. Am J Cardiol. 1993;72:38A-43A.
264. Jacquet L, Evenepoel M, Marenne F, et al. Hemodynamic
effects and safety of sotalol in the prevention of
supraventricular arrhythmias after coronary artery bypass
surgery. J Cardiothorac Vasc Anesth. 1994;8:431-436.
265. Fernandes CM, Daya MR. Sotalol-induced bradycardia
reversed by glucagon. Can Fam Physician. 1995;41:659660, 663-665.
266. Halinen MO, Huttunen M, Paakkinen S, et al.
Comparison of sotalol with digoxin-quinidine for
conversion of acute atrial fibrillation to sinus rhythm
(the Sotalol-Digoxin-Quinidine Trial). Am J Cardiol.
1995;76:495-498.
267. Lee SH, Chen SA, Tai CT, et al. Comparisons of oral
propafenone and sotalol as an initial treatment in
patients with symptomatic paroxysmal atrial fibrillation.
Am J Cardiol. 1997;79:905-908.
268. Beaufort-Krol GC, Bink-Boelkens MT. Sotalol for atrial
tachycardias after surgery for congenital heart disease.
Pacing Clin Electrophysiol. 1997;20:2125-2129.
269. Maisel WH, Kuntz KM, Reimold SC, et al. Risk of
initiating antiarrhythmic drug therapy for atrial
fibrillation in patients admitted to a university hospital.
Ann Intern Med. 1997;127:281-284.
270. Chung MK, Schweikert RA, Wilkoff BL, et al. Is hospital
admission for initiation of antiarrhythmic therapy with
sotalol for atrial arrhythmias required? Yield of inhospital monitoring and prediction of risk for significant
arrhythmia complications. J Am Coll Cardiol.
1998;32:169-176.
271. Gomes JA, Ip J, Santoni-Rugiu F, Mehta D, et al. Oral d,l
sotalol reduces the incidence of postoperative atrial
fibrillation in coronary artery bypass surgery patients: a
randomized, double-blind, placebo-controlled study. J
Am Coll Cardiol. 1999;34:334-339.
272. Matsuura K, Takahara Y, Sudo Y, et al. Effect of Sotalol in
the prevention of atrial fibrillation following coronary
CHAPTER 25 • Supraventricular Arrhythmias
273.
274.
275.
276.
277.
278.
279.
280.
281.
282.
283.
284.
285.
286.
287.
288.
289.
290.
291.
292.
artery bypass grafting. Jpn J Thorac Cardiovasc Surg.
2001;49:614-617.
Celiker A, Ayabakan C, Ozer S, et al. Sotalol in treatment
of pediatric cardiac arrhythmias. Pediatr Int. 2001;43:624630.
Auer J, Weber T, Berent R, et al. A comparison between
oral antiarrhythmic drugs in the prevention of atrial
fibrillation after cardiac surgery: the pilot study of
prevention of postoperative atrial fibrillation (SPPAF), a
randomized, placebo-controlled trial. Am Heart J.
2004;147:636-643.
Barreto RS. Effect of intravenously administered
succinylcholine upon cardiac rate and rhythm.
Anesthesiology. 1960;21:401-404.
Craythorne NW, Turndorf H, Dripps RD. Changes in the
pulse rate and rhythm associated with the use of
succinylcholine in anesthetized children. Anesthesiology.
1960;21:465-470.
Perez H. Cardiac arrhythmia after succinylcholine. Anesth
Analg. 1970;49:33-38.
Cozanitis DA, Dundee JW, Khan MM. Comparative study
of atropine and glycopyrrolate on suxamethoniuminduced changes in cardiac rate and rhythm. Br J
Anaesth. 1980;52:291-293.
Lerman J, Chinyanga HM. The heart rate response to
succinylcholine in children: a comparison of atropine
and glycopyrrolate. Can Anaesth Soc J. 1983;30:377-381.
Sørensen M, Engbaek J, Viby-Mogensen J, et al.
Bradycardia and cardiac asystole following a single
injection of suxamethonium. Acta Anaesthesiol Scand.
1984;28:232-235.
Green DW, Bristow AS, Fisher M. Comparison of i.v.
glycopyrrolate and atropine in the prevention of
bradycardia and arrhythmias following repeated doses of
suxamethonium in children. Br J Anaesth. 1984;56:981-985.
Inoue K, Reichelt W. Asystole and bradycardia in adult
patients after a single dose of suxamethonium. Acta
Anaesthesiol Scand. 1986;30:571-573.
Sherman EP, Lebowitz PW, Street WC. Bradycardia
following sufentanil-succinylcholine. Anesthesiology.
1987;66:106.
Rivard JC, Lebowitz PW. Bradycardia after alfentanil:
succinylcholine. Anesth Analg. 1988;67:907.
Baraka A. Severe bradycardia following propofolsuxamethonium sequence. Br J Anaesth. 1988;61:482-483.
Hansen D. Suxamethonium-induced cardiac arrest and
death following 5 days of immobilization. Eur J
Anaesthesiol. 1998;15:240-241.
Pace SA, Fuller FP. Out-of-hospital succinylcholineassisted endotracheal intubation by paramedics. Ann
Emerg Med. 2000;35:568-572.
Sawabe T, Mizuno S, Gondo H, et al. Sinus arrest during
tacrolimus (FK506) and digitalis treatment in a bone
marrow transplant recipient. Transplantation.
1997;64:182-183.
Cox TH, Baillie GM, Baliga P. Bradycardia associated with
intravenous administration of tacrolimus in a liver
transplant recipient. Pharmacotherapy. 1997;17:1328-1330.
Juliusson G, Celsing F, Turesson I, et al. Frequent good
partial remissions from thalidomide including best
response ever in patients with advanced refractory and
relapsed myeloma. Br J Haematol. 2000;109:89-96.
Rajkumar SV, Gertz MA, Lacy MQ, et al. Thalidomide as
initial therapy for early-stage myeloma. Leukemia.
2003;17:775-779.
Kaur A, Yu SS, Lee AJ, et al. Thalidomide-induced sinus
bradycardia. Ann Pharmacother. 2003;37:1040-1043.
475
293. Kropff MH, Lang N, Bisping G, et al. Hyperfractionated
cyclophosphamide in combination with pulsed
dexamethasone and thalidomide (HyperCDT) in primary
refractory or relapsed multiple myeloma. Br J Haematol.
2003;122:607-616.
294. Coutsouvelis J, Corallo CE. Thalidomide-induced
bradycardia and its management. Med J Aust.
2004;180:366-367.
295. Fahdi IE, Gaddam V, Saucedo JF, et al. Bradycardia during
therapy for multiple myeloma with thalidomide. Am J
Cardiol. 2004;93:1052-1055.
296. Palladini G, Perfetti V, Perlini S, et al. The combination
of thalidomide and intermediate-dose dexamethasone is
an effective but toxic treatment for patients with primary
Amyloidosis (AL). Blood. 2005;105:2949-2951.
297. Schütt P, Ebeling P, Buttkereit U, et al. Thalidomide in
combination with dexamethasone for pretreated patients
with multiple myeloma: serum level of soluble
interleukin-2 receptor as a predictive factor for response
rate and for survival. Ann Hematol. 2005;84:594-600.
298. Rajkumar SV, Blood E, Vesole D, et al. Phase III clinical
trial of thalidomide plus dexamethasone compared with
dexamethasone alone in newly diagnosed multiple
myeloma: a clinical trial coordinated by the Eastern
Cooperative Oncology Group. J Clin Oncol. 2006;24:431436.
299. Meyer T, Maier A, Borisow N, et al. Thalidomide causes
sinus bradycardia in ALS. J Neurol 2008;255:587-591.
300. Sanborn SL, Cooney MM, Dowlati A, et al. Phase I trial of
docetaxel and thalidomide: a regimen based on
metronomic therapeutic principles. Invest New Drugs.
2008;26:355-362.
301. Yamaguchi T. Syncope and sinus bradycardia from combined
use of thalidomide and beta-blocker. Pharmacoepidemiol Drug
Saf. 2008;Jul 9 (epub ahead of print).
302. de Faire U, Lundman T. Attempted suicide with
verapamil. Eur J Cardiol. 1977;6:195-198.
303. de Faire U, Lundman T. Serious verapamil poisoning. Br J
Med. 1978;2:1574.
304. Jain SC, Bharadwaj RP, Shukla RK, et al. Severe
hypotension and bradycardia after suicidal consumption
of 100 tablets of verapamil: a case report. Indian Heart J.
1982;34:175-176.
305. Wayne VS, Harper RW, Laufer E, et al. Adverse
interaction between beta-adrenergic blocking drugs and
verapamil: report of three cases. Aust N Z J Med.
1982;12:285-289.
306. Subramanian VB, Bowles MJ, Khurmi NS, et al.
Randomized double-blind comparison of verapamil and
nifedipine in chronic stable angina. Am J Cardiol.
1982;50:696-703.
307. Crump BJ, Holt DW, Vale JA. Lack of response to
intravenous calcium in severe verapamil poisoning.
Lancet. 1982;2:939-940.
308. Leitner RP, Hawker RE, Celermajer JM. Intravenous
verapamil in the treatment of paroxysmal
supraventricular tachycardia in children. Aust Paediatr J.
1983;19:40-44.
309. Radford D. Side effects of verapamil in infants. Arch Dis
Child. 1983;58:465-466.
310. Morris DL, Goldschlager N. Calcium infusion for reversal
of adverse effects of intravenous verapamil. JAMA.
1983;249:3212-3213.
311. Subramanian VB, Bowles MJ, Khurmi NS, et al.
Evaluation of verapamil and high dose nifedipine in
patients with chronic stable angina with objective
methods. Acta Med Scand. 1984;681;61-73.
476
SECTION VI • Drug-Induced Cardiovascular Diseases
312. Passal DB, Crespin FH Jr. Verapamil poisoning in an
infant. Pediatrics. 1984;73:543-545.
313. Zatuchni J. Bradycardia and hypotension after
propranolol HCl and verapamil. Heart Lung. 1985;14:9495.
314. Winniford MD, Fulton KL, Corbett JR, et al. Propranololverapamil versus propranolol-nifedipine in severe angina
pectoris of effort: a randomized, double-blind, crossover
study. Am J Cardiol. 1985;55:281-285.
315. Purnell RJ. Bradycardia after verapamil and spinal
analgesia. Anaesthesia. 1985;40:1245.
316. Pennock GD, Dalton WS, Roeske WR, et al. Systemic
toxic effects associated with high-dose verapamil
infusion and chemotherapy administration. J Natl Cancer
Inst. 1991;83:105-110.
317. Quezado Z, Lippmann M, Wertheimer J. Severe cardiac,
respiratory, and metabolic complications of massive
verapamil overdose. Crit Care Med. 1991;19:436-438.
318. Watson NA, FitzGerald CP. Management of massive
verapamil overdose. Med J Aust. 1991;155:124-125.
319. Arstall MA, Beltrame JF, Mohan P, et al. Incidence of
adverse events during treatment with verapamil for
suspected acute myocardial infarction. Am J Cardiol.
1992;70:1611-1612.
320. Doyon S, Roberts JR. The use of glucagon in a case of
calcium channel blocker overdose. Ann Emerg Med.
1993;2:1229-1233.
321. Howarth DM, Dawson AH, Smith AJ, et al. Calcium
channel blocking drug overdose: an Australian series.
Hum Exp Toxicol. 1994;13:161-166.
322. Ashraf M, Chaudhary K, Nelson J, et al. Massive overdose
of sustained-release verapamil: a case report and review
of literature. Am J Med Sci. 1995;310:258-253.
323. Van Mieghem W, Tits G, Demuynck K, et al. Verapamil
as prophylactic treatment for atrial fibrillation after lung
operations. Ann Thorac Surg. 1996;61:1083-1085.
324. Pahor M, Manto A, Pedone C, et al. Age and severe
adverse drug reactions caused by nifedipine and
verapamil: Gruppo Italiano di Farmacovigilanza nell’
Anziano (GIFA). J Clin Epidemiol. 1996;49:921-928.
325. Brass BJ, Winchester-Penny S, Lipper BL. Massive
verapamil overdose complicated by noncardiogenic
pulmonary edema. Am J Emerg Med. 1996;14:459-461.
326. Yörükoğlu D, Göktug A, Alanoğlu Z, et al. Comparison of
intravenous metoprolol, verapamil and diltiazem on the
attenuation of haemodynamic changes associated with
tracheal extubation. Eur J Anaesthesiol. 1999;16:462-467.
327. Yuan TH, Kerns WP 2nd, Tomaszewski CA, et al. Insulinglucose as adjunctive therapy for severe calcium channel
antagonist poisoning. J Toxicol Clin Toxicol. 1999;37:463474.
328. Kuhlmann U, Schoenemann H, Müller T, et al.
Plasmapheresis in life-threatening verapamil
intoxication. Artif Cells Blood Substit Immobil Biotechnol.
2008;28:429-440.
329. White WB, Johnson MF, Anders RJ, et al. Safety of
controlled-onset extended-release verapamil in middleaged and older patients with hypertension and coronary
artery disease. Am Heart J. 2001;142:1010-1015.
330. Kato M, Dote K, Sasaki S, et al. Intracoronary verapamil
rapidly terminates reperfusion tachyarrhythmias in acute
myocardial infarction. Chest. 2004;126:702-708.
331. Netland PA, Weiss HS, Stewart WC, et al. Cardiovascular
effects of topical carteolol hydrochloride and timolol
maleate in patients with ocular hypertension and
primary open-angle glaucoma: Night Study Group. Am J
Ophthalmol. 1997;123:465-477.
332. Williams T, Ginther WH. Hazard of ophthalmic timolol.
N Engl J Med. 1982;306:1485-1486.
333. Guha S, Dawn B, Dutta G, et al. Bradycardia, reversible
panconduction defect and syncope following selfmedication with a homeopathic medicine. Cardiology.
1999;91:268-271.
334. Chan TY. Aconite poisoning. Clin Toxicol. 2009;47:279285.
335. DiMarco JP, Sellers TD, Berne RM, et al. Adenosine:
electrophysiologic effects and therapeutic use for
terminating paroxysmal supraventricular tachycardia.
Circulation. 1983;68:1254-1263.
336. Tajima T, Muramatsu T, Kanaka S, et al.
Electrophysiologic effects of intravenous adenosine
triphosphate disodium on the paroxysmal
supraventricular tachycardias. Jpn Heart J. 1985;26:33-43.
337. diMarco JP, Sellers TD, Lerman BB, et al. Diagnostic and
therapeutic use of adenosine in patients with
supraventricular tachyarrhythmias. J Am Coll Cardiol.
1985;6:417-425.
338. Watt AH, Routledge PA. Transient bradycardia and
subsequent sinus tachycardia produced by intravenous
adenosine in healthy adult subjects. Br J Clin Pharmacol.
1986;21:533-536.
339. Sylvén C, Jonzon B, Brandt R, et al. Adenosine-provoked
angina pectoris-like pain: time characteristics, influence
of autonomic blockade and naloxone. Eur Heart J.
1987;8:738-743.
340. Sylvén C, Jonzon B, Edlund A. Angina pectoris-like pain
provoked by i.v. bolus of adenosine: relationship to
coronary sinus blood flow, heart rate and blood pressure
in healthy volunteers. Eur Heart J. 1989;10:48-54.
341. Till J, Shinebourne EA, Rigby ML, et al. Efficacy and
safety of adenosine in the treatment of supraventricular
tachycardia in infants and children. Br Heart J.
1989;62:204-211.
342. Rossen JD, Quillen JE, Lopez AG, et al. Comparison of
coronary vasodilation with intravenous dipyridamole
and adenosine. J Am Coll Cardiol. 1991;18:485-491.
343. Abreu A, Mahmarian JJ, Nishimura S, et al. Tolerance and
safety of pharmacologic coronary vasodilation with
adenosine in association with thallium-201 scintigraphy
in patients with suspected coronary artery disease. J Am
Coll Cardiol. 1991;18:730-735.
344. Lee J, Heo J, Ogilby JD, et al. Atrioventricular block
during adenosine thallium imaging. Am Heart J.
1992;123:1569-1574.
345. Anderson TJ, Ryan TJ Jr, Mudge GH, et al. Sinoatrial and
atrioventricular block caused by intracoronary infusion
of adenosine early after heart transplantation. Heart Lung
Transplant. 1993;12:522-524.
346. Toft J, Mortensen J, Hesse B. Risk of atrioventricular
block during adenosine pharmacologic stress testing in
heart transplant recipients. Am J Cardiol. 1998;82:696697.
347. Alkoutami GS, Reeves WC, Movahed A. The safety of
adenosine pharmacologic stress testing in patients with
first-degree atrioventricular block in the presence and
absence of atrioventricular blocking medications. J Nucl
Cardiol. 1999;6:495-497.
348. Alkoutami GS, Reeves WC, Movahed A. The frequency of
atrioventricular block during adenosine stress testing in
young, middle-aged, young-old, and old-old adults. Am J
Geriatr Cardiol. 2001;10:159-161.
349. Camaiti A, Pieralli F, Olivotto I, et al. Prospective
evaluation of adenosine-induced proarrhythmia in the
emergency room. Eur J Emerg Med. 2001;8:99-105.
CHAPTER 25 • Supraventricular Arrhythmias
350. Treuth MG, Reyes GA, He ZX, et al. Tolerance and
diagnostic accuracy of an abbreviated adenosine infusion
for myocardial scintigraphy: a randomized, prospective
study. J Nucl Cardiol. 2001;8:548-554.
351. Makaryus JN, Catanzaro JN, Friedman ML, et al.
Persistent second-degree atrioventricular block following
adenosine infusion for nuclear stress testing. J Cardiovasc
Med. 2008;9:304-307.
352. Ilyas M. Intermittent atrioventricular block after
propranolol. N Engl J Med. 1972;286:376-377.
353. Zeis PM, Thanopoulos B, Pierroutsakos IN, et al.
Complete atrioventricular block associated with
propranolol therapy. J Pediatr. 1981;98:326-327.
354. García-Rubira JC, Pavón M, Romero-Chacón D. Syncope
induced by propranolol in hypertrophic cardiomyopathy.
Int J Cardiol. 1991;31:358-361.
355. Kelkar PN. Atenolol induced high grade AV block. J Assoc
Physicians India. 1998;46:748-751.
356. Krum H, Shusterman N, MacMahon S, et al. Efficacy and
safety of carvedilol in patients with chronic heart failure
receiving concomitant amiodarone therapy:
Australia/New Zealand Heart Failure Research
Collaborative Group. J Cardiac Fail. 1998;4:281-288.
357. Nägele H, Bohlmann M, Eck U, et al. Combination
therapy with carvedilol and amiodarone in patients with
severe heart failure. Eur J Heart Fail. 2000;2:71-79.
358. Ruiz-Ruiz FJ, Perez-Calvo JI, Sanjuan-Cuartero R. Topical
beta blockers and atrioventricular block in the elderly. J
Postgrad Med. 2002;48:327-328.
359. Zeltser D, Justo D, Halkin A, et al. Drug-induced
atrioventricular block: prognosis after discontinuation of
the culprit drug. J Am Coll Cardiol. 2004;44:105-108.
360. Tarantini L, Cioffi G, Opasich C, et al. Pre-discharge
initiation of beta-blocker therapy in elderly patients
hospitalized for acute decompensation of chronic heart
failure: an effective strategy for the implementation of
beta-blockade in heart failure. Ital Heart J. 2004;5:441449.
361. Onalan O, Cumurcu BE, Bekar L. Complete
atrioventricular block associated with concomitant use of
metoprolol and paroxetine. Mayo Clin Proc. 2008;83:595599.
362. Morady F, Scheinman MM, Shen E, et al. Intravenous
amiodarone in the acute treatment of recurrent
symptomatic ventricular tachycardia. Am J Cardiol.
1983;51:156-159.
363. Mangiardi L, Gaita F, Brun S, et al. Atrioventricular
block complicating amiodarone-induced
hypothyroidism in a patient with pre-excitation and
rate-dependent bilateral bundle branch block. J Am Coll
Cardiol. 1986;7:180-184.
364. Raviña T, Gutierrez J. Amiodarone-induced AV block and
ventricular standstill: a forme fruste of an idiopathic long
QT syndrome. Int J Cardiol. 2000;75:105-108.
365. McMahon CJ, Laird WP, Fenrich AL. Amiodaroneinduced 2 to 1 atrioventricular block in association with
prolongation of the QT interval. Cardiol Young.
2003;13:305-307.
366. Saul JP, Scott WA, Brown S, et al. Intravenous
amiodarone for incessant tachyarrhythmias in children:
a randomized, double-blind, antiarrhythmic drug trial.
Circulation. 2005;112:3470-3477.
367. Smith RB, Rusbatch BJ. Amitriptyline and heart block. Br
Med J. 1967;3:311.
368. Lappa A, Castagna A, Imperiale C, et al. Near fatal case of
atrio-ventricular block induced by amitriptyline at
therapeutic dose. Intensive Care Med. 2000;26:1399.
477
369. Timour Q, Freysz M, Couzon P, et al. Possible role of drug
interactions in bupivicaine-induced problems related to
intraventricular conduction disorders. Reg Anesth.
1990;15:180-185.
370. Shen CL, Ho YY, Hung YC, et al. Arrhythmias during
spinal anesthesia for Cesarean section. Can J Anaesth.
2000;47:393-397.
371. Beermann B, Edhag O, Vallin H. Advanced heart block
aggravated by carbamazepine. Br Heart J. 1975;37:668671.
372. Ladefoged SD, Møgelvang JC. Total atrioventricular block
with syncopes complicating carbamazepine therapy. Acta
Med Scand. 1982;212:185-186.
373. Boesen F, Andersen EB, Jensen EK, et al. Cardiac
conduction disturbances during carbamazepine therapy.
Acta Neurol Scand .1983;68:49-52.
374. Labrecque J, Coté MA, Vincent P. Carbamazepineinduced atrioventricular block. Am J Psychiatry.
1992;149:572-573.
375. Hantson P, Ilunga K, Martin N, et al. Cardiac conduction
abnormalities during carbamazepine therapy for
neuralgia following Guillain-Barré syndrome. Acta Neurol
Belg. 1993;93:40-43.
376. Ide A, Kamijo Y. Intermittent complete atrioventricular
block after long term low-dose carbamazepine therapy
with a serum concentration less than the therapeutic
level. Intern Med. 2007;46:627-629.
377. Guedira N, Hajjaj-Hassouni N, Srairi JE, et al. Thirddegree atrioventricular block in a patient under
chloroquine therapy. Rev Rhum Engl Ed. 1998;65:58-62.
378. Reuss-Borst M, Berner B, Wulf G, et al. Complete heart
block as a rare complication of treatment with
chloroquine. J Rheumatol. 1999;26:1394-1395.
379. Teixeira RA, Martinelli Filho M, Benvenuti LA, et al.
Cardiac damage from chronic use of chloroquine: a case
report and review of the literature. Arg Bras Cardiol.
2002;79:85-88.
380. Tordjman T, Korzets A, Kotas R, et al. Complete
atrioventricular block and long-term cimetidine therapy.
Arch Intern Med. 1994;144:861.
381. Gambassi G, Incalzi RA, Gemma A. Atrioventricular
blocks associated with citalopram. Am J Geriatr Psychiatry.
2005;13:918-919.
382. Williams PL, Krafcik JM, Potter BB, et al. Cardiac toxicity
of clonidine. Chest.1977;72:784-785.
383. Abiuso P, Abelow G. Atrioventricular dissociation in a
patient receiving clonidine. JAMA. 1978;240:108-109.
384. Ciaccheri M, Dolara A, Manetti A, et al. A-V block by an
overdose of Clonidine. Acta Cardiol. 1983;38:233-236.
385. Jaffe R, Livshits T, Bursztyn M. Adverse interaction
between clonidine and verapamil. Ann Pharmacother.
1994;28:881-883.
386. Om A, Ellenbogen KA, Vetrovec GW. Cocaine-induced
bradyarrhythmias. Am Heart J. 1992;124:232-234.
387. Smith TW, Haber E. Digoxin intoxication: the
relationship of clinical presentation to serum digoxin
concentration. J Clin Invest. 1970;49:2377-2386.
388. Duke M. Atrioventricular block due to accidental digoxin
ingestion treated with atropine. Am J Dis Child.
1972;124:754-756.
389. Smith TW, Haber E, Yeatman L, et al. Reversal of
advanced digoxin intoxication with Fab fragments of
digoxin-specific antibodies. N Engl J Med. 1976;294:797800.
390. Chan W, Ikram H. A case of self-induced digoxin
poisoning: with His bundle studies of the site of heart
block. N Z Med J. 1976;84:443-446.
478
SECTION VI • Drug-Induced Cardiovascular Diseases
391. Rostás L, Tarján J. Wenckebach type H-V block induced
by digitalis intoxication. Z Kardiol. 1980;69:779-781.
392. Goren C, Denes P. The role of Holter monitoring in
detecting digitalis-provoked arrhythmias. Chest.
1981;79:555-558.
393. Rossi R, Leititis JU, Hagel KJ, et al. Severe digoxin
intoxication in a child treated by infusion of digoxinspecific Fab-antibody-fragments. Eur J Pediatr.
1984;142:138-140.
394. Smolarz A, Roesch E, Lenz E, et al. Digoxin specific
antibody (Fab) fragments in 34 cases of severe digitalis
intoxication. J Toxicol Clin Toxicol. 1985;23:327-340.
395. Moorman JR. Digitalis toxicity at Duke Hospital, 19731984. South Med J. 1985;78:561-564.
396. Lai KN, Swaminathan R, Pun CO, et al. Hemofiltration in
digoxin overdose. Arch Intern Med. 1986;146:1219-1220.
397. Lewander WJ, Gaudreault P, Einhorn A, et al. Acute
pediatric digoxin ingestion: a ten-year experience. Am J
Dis Child. 1986;140:770-773.
398. Kearns GL, Moss MM, Clayton BD, et al.
Pharmacokinetics and efficacy of digoxin specific Fab
fragments in a child following massive digoxin overdose.
J Clin Pharmacol. 1989;29:901-908.
399. Woolf AD, Wenger T, Smith TW, et al. The use of
digoxin-specific Fab fragments for severe digitalis
intoxication in children. N Engl J Med. 1992;326:17391744.
400. Wofford JL, Hickey AR, Ettinger WH, et al. Lack of agerelated differences in the clinical presentation of digoxin
toxicity. Arch Intern Med. 1992;152:2261-2264.
401. Abad-Santos F, Carcas AJ, Ibáñez C, et al. Digoxin level
and clinical manifestations as determinants in the
diagnosis of digoxin toxicity. Ther Drug Monit.
2000;22:163-168.
402. Spodick DH. Well concealed atrial tachycardia with
Wenckebach (Mobitz I) atrioventricular block: digitalis
toxicity. Am J Geriatr Cardiol. 2001;10:59.
403. Hossack KF. Conduction abnormalities due to diltiazem.
N Engl J Med. 1982;307:953-954.
404. Bessho F, Kinumaki H, Kobayashi M, et al. Treatment of
children with refractory acute lymphocytic leukemia
with vincristine and diltiazem. Med Pediatr Oncol.
1985;13:199-202.
405. Ramoska EA, Spiller HA, Winter M, et al. A one-year
evaluation of calcium channel blocker overdoses: toxicity
and treatment. Ann Emerg Med. 1993;22:196-200.
406. Shammas RL, DeAntonio HJ, Movahed A. High-grade
atrioventricular block during dipyridamole stress testing.
Int J Cardiol. 1994;46:279-282.
407. Preumont N, Velez Roa S, Berkenboom G, et al. Transient
second-degree heart block during dipyridamole myocardial
perfusion imaging. Acta Cardiol. 1998;53:275-277.
408. Alakhras M, Baldari D, El-Sherif N, et al. Location and
clinical implications of high-degree atrioventricular block
during dipyridamole infusion: a case report. Ann
Noninvasive Electrocardiol. 2002;7:174-176.
409. Camm J, Ward D, Spurrell RA. The effect of intravenous
disopyramide phosphate on recurrent paroxysmal
tachycardias. Br J Clin Pharmacol. 1979;8:441-449.
410. Rothman MT. Prolonged QT interval, atrioventricular
block, and torsade de pointes after antiarrhythmic
therapy. Br J Med. 1980;280:922-933.
411. Bergfeldt L, Rosenqvist M, Vallin H, et al. Disopyramide
induced second and third degree atrioventricular block
in patients with bifascicular block: an acute stress test to
predict atrioventricular block progression. Br Heart J.
1985;53:328-334.
412. Suleyman T, Tevfik P, Abdulkadir G, et al. Complete
atrioventricular block and ventricular tachyarrhythmia
associated with donepezil. Emerg Med J. 2006;23:641-642.
413. Combeer A. Complete atrioventricular block following
etomidate. Eur J Anaesthesiol. 2007;24:1067-1068.
414. Hanley NA, Bourke JP, Gascoigne AD. Survival in a case
of life-threatening flecainide overdose. Intensive Care Med.
1998;24:740-742.
415. LaRusso P, Jessup SA, Rogers FJ, et al. Sinoatrial and
atrioventricular dysfunction associated with the use of
guanabenz acetate. Can J Cardiol. 1988;4:146-148.
416. Comin-Colet J, Sanchez-Corral MA, Alegre-Sancho JJ, et
al. Complete heart block in an adult with systemic lupus
erythematosus and recent onset of hydroxychloroquine
therapy. Lupus. 2001;10:59-62.
417. Kantor SJ, Bigger JT Jr, Glassman AH, et al. Imipramineinduced heart block: a longitudinal case study. JAMA.
1975;231:1364-1366.
418. Martin CI. Imipramine-induced heart block. JAMA.
1975;234:383.
419. Ibanez L, Laporte JR, Carne X. Adverse drug reactions
leading to hospital admission. Drug Saf. 1991;6:450-459.
420. Wheeler DW, Liew TV, Bailey AR. Peri-operative
atrioventricular block as a result of chemotherapy with
epirubicin and paclitaxel. Anaesthesia. 2007;62:186-189.
421. Antoniou T, Gough KA. Early-onset pentamidineassociated second-degree heart block and sinus
bradycardia: case report and review of the literature.
Pharmacotherapy. 2005;25:899-903.
422. Burton BT, Rice M, Schmertzler LE. Atrioventricular block
following overdose of decongestant cold medication. J
Emerg Med. 1985;2:415-419.
423. Woo OF, Benowitz NL, Bialy FW, et al. Atrioventricular
conduction block caused by phenylpropanolamine.
JAMA. 1985;253:2646-2647.
424. Libersa C, Caron J, Pladys A, et al. Propafenone versus
disopyramide: a double-blind randomized crossover trial
in patients presenting chronic ventricular arrhythmias.
Clin Cardiol. 1987;10:405-410.
425. Kerns W 2nd, English B, Ford M. Propafenone overdose.
Ann Emerg Med. 1994;24:98-103.
426. Janousek J, Paul T. Safety of oral propafenone in the
treatment of arrhythmias in infants and children
(European retrospective multicenter study): Working
Group on Pediatric Arrhythmias and Electrophysiology
of the Association of European Pediatric Cardiologists.
Am J Cardiol. 1998;81:1121-1124.
427. James MF, Reyneke CJ, Whiffler K. Heart block
following propofol: a case report. Br J Anaesth.
1989;62:213-215.
428. Ganansia MF, Francois TP, Ormezzano X, et al.
Atrioventricular Mobitz I block during propofol
anesthesia for laparoscopic tubal ligation. Anesth Analg.
1989;69:524-525.
429. Sochala C, Deenen D, Ville A, et al. Heart block
following propofol in a child. Paediatr Anaesth.
1999;9:349-351.
430. Gehi A, Benatar M, Langberg J. Treatment of
pyridostigmine-induced AV block with hyoscyamine in a
patient with myasthenia gravis. J Cardiovasc
Electrophysiol. 2008;19:214-216.
431. Pfammatter JP, Paul T, Lehmann C, et al. Efficacy and
proarrhythmia of oral sotalol in pediatric patients. J Am
Coll Cardiol. 1995;26:1002-1007.
432. Donatini B, Le Blaye I, Krupp P. Transient cardiac pacing
is insufficiently used to treat arrhythmia associated with
thioridazine. Cardiology. 1992;81:340-341.
CHAPTER 25 • Supraventricular Arrhythmias
433. Hulisz DT, Dasa SL, Black LD, et al. Complete heart block
and torsade de pointes associated with thioridazine
poisoning. Pharmacotherapy. 1994;14:239-245.
434. de Faire U, Lundman T. Attempted suicide with
verapamil. Eur J Cardiol. 1977;6:195-198.
435. Chimienti M, Previtali M, Medicia A, et al. Acute
verapamil poisoning: successful treatment with
epinephrine. Clin Cardiol. 1982;5:219-222.
436. Perrot B, Danchin N, Terrier de la Chaise A. Verapamil: a
cause of sudden death in a patient with hypertrophic
cardiomyopathy. Br Heart J. 1984;51:352-354.
437. Danish Multicenter Study Group on Verapamil in
Myocardial Infarction. Verapamil in acute myocardial
infarction. Am J Cardiol. 1984;54:24E-28E.
438. Spiller HA, Meyers A, Ziemba T, et al. Delayed onset of
cardiac arrhythmias from sustained-release verapamil.
Ann Emerg Med. 1991;20:201-203.
439. Pritza DR, Bierman MH, Hammeke MD. Acute toxic
effects of sustained-release verapamil in chronic renal
failure. Arch Intern Med. 1991;151:2081-2084.
440. Ramoska EA, Spiller HA, Winter M, et al. A one-year
evaluation of calcium channel blocker overdoses: toxicity
and treatment. Ann Emerg Med. 1993;22:196-200.
441. Barrow PM, Houston PL, Wong DT. Overdose of
sustained-release verapamil. Br J Anaesth. 1994;72:361365.
442. Martín-Gago J, Pascual J, Rodríguez-Palomares JR, et al.
Complete atrioventricular blockade secondary to
conventional-release verapamil in a patient on
hemodialysis. Nephron. 1999;83:89-90.
443. Goldschmidt N, Azaz-Livshits T, Gotsman, et al.
Compound cardiac toxicity of oral erythromycin and
verapamil. Ann Pharmacother. 2001;35:1396-1399.
444. Werner GS, Lang K, Kuehnert H, et al. Intracoronary
verapamil for reversal of no-reflow during coronary
angioplasty for acute myocardial infarction. Catheter
Cardiovasc Interv. 2002;57:444-451.
445. Vijayalakshmi K, Whittaker VJ, Kunadian B, et al.
Prospective, randomised, controlled trial to study the
effect of intracoronary injection of verapamil and
adenosine on coronary blood flow during percutaneous
coronary intervention in patients with acute coronary
syndromes. Heart. 2006;92:1278-1284.
446. Letavernier E, Couzi L, Delmas Y, et al. Verapamil and
mild hyperkalemia in hemodialysis patients: a
potentially hazardous association. Hemodial Int.
2006;10:170-172.
447. Cohen AS, Matharu MS, Goadsby PJ.
Electrocardiographic abnormalities in patients with
cluster headache on verapamil therapy. Neurology.
2007;69:668-675.
448. Essebag V, Hadjis T, Platt RW, et al. Amiodarone and the
risk of bradyarrhythmia requiring permanent pacemaker
in elderly patients with atrial fibrillation and prior
myocardial infarction. J Am Coll Cardiol. 2003;41:249254.
449. Lee SH, Chang CM, Lu MJ, et al. Intravenous
amiodarone for prevention of atrial fibrillation after
coronary artery bypass grafting. Ann Thorac Surg.
2000;70:157-161.
450. Cotter G, Blatt A, Kaluski E, et al. Conversion of recent
onset paroxysmal atrial fibrillation to normal sinus
rhythm: the effect of no treatment and high-dose
amiodarone: a randomized, placebo-controlled study. Eur
Heart J. 1999;20:1833-1842.
451. Ma S, Long JP. Hypotensive and bradycardiac responses
to reflex sympathetic inhibition produced by
452.
453.
454.
455.
456.
457.
458.
459.
460.
461.
462.
463.
464.
465.
466.
467.
468.
479
nitroglycerin in rats with sinoaortic deafferentation. J
Cardiovasc Pharmacol. 1993;21:136-143.
Bernstein AD, Parsonnet V. Survey of cardiac pacing in
the United States in 1989. Am J Cardiol. 1992;69:331-338.
Melenovsky V, Wichterle D, Malik J, et al. Nitroglycerin
induced syncope occurs in subjects with delayed phase
shift of baroreflex action. PACE. 2002;25:828-832.
Flockhart DA. Drug Interactions: Cytochrome P450 Drug
Interaction Table. Available at:
http://medicine.iupui.edu/flockhart/table.htm. Accessed
July 20, 2009.
Knabb RM, Gidday GM, Ely SW. Effects of dipyridamole
on myocardial adenosine on active hyperemia. Am J
Physiol. 1984;247:804-810.
Fenster PE, White NW Jr, Hanson CD. Pharmacokinetic
evaluation of the digoxin-amiodarone interaction. J Am
Coll Cardiol. 1985;5:108-112.
Klein HO, Lang R, Weiss E, et al. The influence of
verapamil on serum digoxin concentration. Circulation.
1982;65:998-1003.
Chen TS, Friedman HS. Alteration of digoxin
pharmacokinetics by a single dose of quinidine. JAMA.
1980;244:669-672.
Sprague DH. Severe bradycardia after neostigmine in a
patient taking propranolol to control paroxysmal atrial
tachycardia. Anesthesiology. 1975;42:208-210.
Essebag V, Reynolds MR, Hadjis T, et al. Sex differences in
the relationship between amiodarone use and the need
for permanent pacing in patients with atrial fibrillation.
Arch Intern Med. 2007;167:1648-1653.
Surawicz B. Factors affecting tolerance to digoxin. J Am
Coll Cardiol. 1985;5:69A-81A.
The American Heart Association in Collaboration with
the International Liaison Committee on Resuscitation
(ILCOR). Guidelines 2000 for cardiopulmonary
resuscitation and emergency cardiovascular care: an
international consensus on science. Circulation. 2000;
102(suppl I):I112-I128.
Salhanick SD, Shannon MW. Management of calcium
channel antagonist overdose. Drug Saf. 2003;26:65-79.
Zimmerman JL. Poisonings and overdoses in the
intensive care unit: general and specific management
issues. Crit Care Med. 2003:31:2794-2801.
Epstein AE, DiMarco JP, Ellenbogen KA, et al.
ACC/AHA/HRS 2008 guidelines for device-based therapy
of cardiac rhythm abnormalities: executive summary: a
report of the American College of Cardiology/American
Heart Association Task Force on Practice Guidelines
(Writing Committee to Revise the ACC/AHA/ NASPE
2002 Guideline Update for Implantation of Cardiac
Pacemakers and Antiarrhythmia Devices). Heart Rhythm.
2008;5:934-955.
Smith SC, Allen J, Blair SN, et al. AHA/ACC guidelines
for secondary prevention for patients with coronary and
other atherosclerotic vascular disease: 2006 update.
Circulation. 2006;113:2363-2372.
Hunt SA, Abraham WT, Chin MH, et al. 2009 Focused
update incorporated into the ACC/AHA 2005 Guidelines
for the Diagnosis and Management of Heart Failure in
Adults A Report of the American College of Cardiology
Foundation/American Heart Association Task Force on
Practice Guidelines Developed in Collaboration With the
International Society for Heart and Lung
Transplantation. J Am Coll Cardiol. 2009;53:e1-e90.
Belhassen B, Pelleg A, Shoshani D, et al. Atrial fibrillation
induced by adenosine triphosphate. Am J Cardiol.
1984;53:1405-1406.
480
SECTION VI • Drug-Induced Cardiovascular Diseases
469. Garratt CJ, Antoniou A, Griffith MJ, et al. Use of
intravenous adenosine in sinus rhythm as a diagnostic
test for latent preexcitation. Am J Cardiol. 1990;65:868873.
470. Meurer MK. A 21-year-old woman with rapid atrial
fibrillation after adenosine administration. J Emerg Nurs.
1991;17:135-136.
471. Cowell RP, Paul VE, Ilsley CD. Haemodynamic
deterioration after treatment with adenosine. Br Heart J.
1994;71:569-571.
472. Crosson JE, Etheridge SP, Milstein S, et al. Therapeutic
and diagnostic utility of adenosine during tachycardia
evaluation in children. Am J Cardiol. 1994;74:155-160.
473. Tebbenjohanns J, Pfeiffer D, Schumacher B, et al.
Intravenous adenosine during atrioventricular reentrant
tachycardia: induction of atrial fibrillation with rapid
conduction over an accessory pathway. Pacing Clin
Electrophysiol. 1995;18:743-746.
474. Silverman AJ, Machado C, Baga JJ, et al. Adenosineinduced atrial fibrillation. Am J Emerg Med. 1996;14:300301.
475. Strickberger SA, Man KC, Daoud EG, et al. Adenosineinduced atrial arrhythmia: a prospective analysis. Ann
Intern Med. 1997;127:417-422.
476. Kaplan IV, Kaplan AV, Fisher JD. Adenosine induced
atrial fibrillation precipitating polymorphic
ventricular tachycardia. Pacing Clin Electrophysiol.
2000;23:140-141.
477. Israel C, Klingenheben T, Grönefeld G, et al. Adenosineinduced atrial fibrillation. J Cardiovasc Electrophysiol.
2000;11:825.
478. Camaiti A, Pieralli F, Olivotto I, et al. Prospective
evaluation of adenosine-induced proarrhythmia in the
emergency room. Eur J Emerg Med. 2001;8:99-105.
479. Kaltman JR, Tanel RE, Shah MJ, et al. Induction of atrial
fibrillation after the routine use of adenosine. Pediatr
Emerg Care. 2006;22:113-115.
480. Cummings M, Raza J, Movahed A. Atrial fibrillation
during adenosine pharmacologic stress testing. J Nucl
Cardiol. 2006;13:576-581.
481. Stuber T, Lim PB, O’Neill MD, et al. Ultra rapid local
activity in adenosine-induced atrial fibrillation. J
Cardiovasc Electrophysiol. 2008;19:566-567.
482. Kanei Y, Hanon S, Van-Tosh A, et al. Adenosine-induced
atrial fibrillation during pharmacologic stress testing:
report of eight cases and review of the literature. Int J
Cardiol. 2008;129:e15-e17.
483. Poukkula A, Korhonen UR, Huikuri H, et al.
Theophylline and salbutamol in combination in patients
with obstructive pulmonary disease and concurrent heart
disease: effect on cardiac arrhythmias. J Intern Med.
1989;226:229-234.
484. Breeden CC, Safirstein BH. Spacer-induced atrial
fibrillation. N J Med. 1990;87:113-114.
485. Breeden CC, Safirstein BH. Albuterol and spacer-induced
atrial fibrillation. Chest. 1990;98:762-763.
486. Ettinger PO, Wu CF, De La Cruz C Jr, et al. Arrhythmias
and the “Holiday Heart”: alcohol-associated cardiac
rhythm disorders. Am Heart J. 1978;95:555-562.
487. Thornton JR. Atrial fibrillation in healthy non-alcoholic
people after an alcoholic binge. Lancet.
1984;2(8410):1013-1015.
488. Nissen MB, Lemberg L. The “Holiday Heart” syndrome.
Heart Lung. 1984;13:89-92.
489. Rich EC, Siebold C, Campion B. Alcohol-related acute
atrial fibrillation: a case-control study and review of 40
patients. Arch Intern Med. 1985;145:830-833.
490. Watt AH. Atrial fibrillation and alcohol. Lancet.
1985;1(8421):162.
491. Sipila R. Atrial fibrillation precipitated by alcohol. Lancet.
1985;1(8425):391-392.
492. Koskinen P, Kupari M, Leinonen H, et al. Alcohol and
new onset atrial fibrillation: a case-control study of a
current series. Br Heart J. 1987;57:468-473.
493. Koskinen P, Kupari M, Leinonen H. Role of alcohol in
recurrences of atrial fibrillation in persons < 65 years of
age. Am J Cardiol. 1990;66:954-958.
494. Maki T, Toivonen L, Koskinen P, et al. Effect of ethanol
drinking, hangover, and exercise on adrenergic activity
and heart rate variability in patients with a history of
alcohol-induced atrial fibrillation. Am J Cardiol.
1998;82:317-322.
495. Conen D, Tedrow UB, Cook NR, et al. Alcohol
consumption and risk of incident atrial fibrillation in
women. JAMA. 2008;300:2489-2496.
496. Cummings SR, Schwartz AV, Black DM. Alendronate and
atrial fibrillation. N Engl J Med. 2007;356:1895-1896.
497. Heckbert SR, Li G, Cummings SR, et al. Use of
alendronate and risk of incident atrial fibrillation in
women. Arch Intern Med. 2008;168:826-831.
498. Tai CT, Chiang CE, Lee SH, et al. Persistent atrial flutter
in patients treated for atrial fibrillation with amiodarone
and propafenone: electrophysiologic characteristics,
radiofrequency catheter ablation, and risk prediction. J
Cardiovasc Electrophysiol. 1999;10:1180-1187.
499. Reithmann C, Hoffmann E, Spitzlberger G, et al.
Catheter ablation of atrial flutter due to amiodarone
therapy for paroxysmal atrial fibrillation. Eur Heart J.
2000;21:565-572.
500. Mattioli AV, Bonatti S, Zennaro M, et al. Effect of coffee
consumption, lifestyle and acute life stress in the
development of acute lone atrial fibrillation. J Cardiovasc
Med. 2008;9:794-798.
501. Doiuchi J, Hamada M, Ochi T, et al. Adverse effects of
atrial fibrillation and syncope induced by calciumchannel blockers in hypertrophic cardiomyopathy. Clin
Cardiol. 1985;8:176-179.
502. Baig MW, Sheard K, Thorley PJ, et al. The use of
dobutamine stress thallium scintigraphy in the diagnosis
of syndrome X. Postgrad Med J. 1992;68(suppl 2):S20-S24.
503. Poldermans D, Fioretti PM, Boersma E, et al. Safety of
dobutamine-atropine stress echocardiography in patients
with suspected or proven coronary artery disease. Am J
Cardiol. 1994;73:456-459.
504. Poldermans D, Fioretti PM, Boersma E, et al.
Dobutamine-atropine stress echocardiography in elderly
patients unable to perform an exercise test:
hemodynamic characteristics, safety, and prognostic
value. Arch Intern Med. 1994;154:2681-2686.
505. Wirtz CE. Sustained atrial fibrillation after dobutamine
stress echocardiography in an older patient with left
atrial enlargement. West J Med. 1995;162:268-269.
506. Cornel JH, Balk AH, Boersma E, et al. Safety and
feasibility of dobutamine-atropine stress
echocardiography in patients with ischemic left
ventricular dysfunction. J Am Soc Echocardiogr. 1996;9:2732.
507. Yeo TC, Ng WL, Ling LH, et al. Dobutamine stress
echocardiography in the elderly Asian patients. Ann Acad
Med Singapore. 1997;26:165-167.
508. Pezzano A, Gentile F, Mantero A, et al. RITED (Registro
Italiano Test Eco-Dobutamina): side effects and
complications of echo-dobutamine stress test in 3041
examinations. G Ital Cardiol. 1998;28:102-111.
CHAPTER 25 • Supraventricular Arrhythmias
509. Elhendy A, van Domburg RT, Bax JJ, et al. Safety,
hemodynamic profile, and feasibility of dobutamine
stress technetium myocardial perfusion single-photon
emission CT imaging for evaluation of coronary artery
disease in the elderly. Chest. 2000;117:649-656.
510. Coisne D, Donal E, Torremocha F, et al. Dobutamine
stress echocardiography response of asymptomatic
patients with diabetes. Echocardiography. 2001;18:373379.
511. Cortigiani L, Zanetti L, Bigi R, et al. Safety and feasibility
of dobutamine and dipyridamole stress
echocardiography in hypertensive patients. J Hypertens.
2002;20:1423-1429.
512. Wahl A, Paetsch I, Gollesch A, et al. Safety and feasibility
of high-dose dobutamine-atropine stress cardiovascular
magnetic resonance for diagnosis of myocardial
ischaemia: experience in 1000 consecutive cases. Eur
Heart J. 2004;25:1230-1236.
513. Onorati F, Renzulli A, De Feo M, et al. Perioperative
enoximone infusion improves cardiac enzyme release
after CABG. J Cardiothorac Vasc Anesth. 2004;18:409-414.
514. Carasso S, Sandach A, Kuperstein R, et al. Atrial
fibrillation in dobutamine stress echocardiography. Int J
Cardiol. 2006;111:53-58.
515. Mebazaa A, Nieminen MS, Packer M, et al. Levosimendan
vs dobutamine for patients with acute decompensated
heart failure: the SURVIVE Randomized Trial. JAMA.
2007;297;1883-1891.
516. Kane GC, Hepinstall MJ, Kidd GM, et al. Safety of stress
echocardiography supervised by registered nurses: results
of a 2-year audit of 15,404 patients. J Am Soc Echocardiogr.
2008;21:337-341.
517. Feld GK, Chen PS, Nicod P, et al. Possible atrial
proarrhythmic effects of class 1C antiarrhythmic drugs.
Am J Cardiol. 1990;66:378-383.
518. Nabar A, Rodriguez LM, Timmermans C, et al. Class IC
antiarrhythmic drug induced atrial flutter:
electrocardiographic and electrophysiological findings
and their importance for long term outcome after right
atrial isthmus ablation. Heart. 2001;85:424-429.
519. Roberge RJ, Martin TG. Mixed fluoxetine/loxapine
overdose and atrial flutter. Ann Emerg Med. 1994;23:586590.
520. O’Driscoll BR. Supraventricular tachycardia caused by
nebulised ipratropium bromide. Thorax. 1989;44:312.
521. McDermott MF, Nasr I, Rydman RJ, et al. Comparison of
two regimens of beta-adrenergics in acute asthma. J Med
Syst. 1999;23:269-279.
522. Ueda N, Yoshikawa T, Chihara M, et al. Atrial fibrillation
following methylprednisolone pulse therapy. Pediatr
Nephrol. 1988;2:29-31.
523. McLuckie AE, Savage RW. Atrial fibrillation following
pulse methylprednisolone therapy in an adult. Chest.
1993;104:622-623.
524. Moretti R, Torre P, Antonello RM, et al. Recurrent atrial
fibrillation associated with pulse administration of high
doses of methylprednisolone: a possible prophylactic
treatment. Eur J Neurol. 2000;7:130.
525. Aslam AK, Vasavada BC, Sacchi TJ, et al. Atrial fibrillation
associated with systemic lupus erythematosus and use of
methylprednisolone. Am J Ther. 2001;8:303-305.
526. Dogukan A, Ilkay E, Poyrazoglu OK, et al. Atrial
fibrillation due to oral methylprednisolone in a patient
with membranoproliferative glomerulonephritis. Acta
Medica. 2008;51:63-64.
527. Wright EM, Sherry KM. Clinical and haemodynamic
effects of milrinone in the treatment of low cardiac
528.
529.
530.
531.
532.
533.
534.
535.
536.
537.
538.
539.
540.
541.
542.
543.
544.
545.
546.
481
output after cardiac surgery. Br J Anaesth. 1991;67:585590.
Feneck RO. Intravenous milrinone following cardiac
surgery: I. Effects of bolus infusion followed by variable
dose maintenance infusion: the European Milrinone
Multicentre Trial Group. J Cardiovasc Thorac Anesth.
1992;6:554-562.
Cuffe MS, Califf RM, Adams KF Jr, et al. Outcomes of a
Prospective Trial of Intravenous Milrinone for
Exacerbations of Chronic Heart Failure (OPTIME-CHF)
Investigators: short-term intravenous milrinone for acute
exacerbation of chronic heart failure: a randomized
controlled trial. JAMA. 2002;287:1541-1547.
Fleming GA, Murray KT, Yu C, et al. Milrinone use is
associated with postoperative atrial fibrillation after
cardiac surgery. Circulation. 2008;118:1619-1625.
Zingler VC, Nabauer M, Jahn K, et al. Assessment of
potential cardiotoxic side effects of mitoxantrone in
patients with multiple sclerosis. Eur Neurol. 2005;54:2833.
Kasinath NS, Malak O, Tetzlaff J. Atrial fibrillation after
ondansetron for the prevention and treatment of
postoperative nausea and vomiting: a case report. Can J
Anaesth. 2003;50:229-231.
Havrilla PL, Kane-Gill SL, Verrico MM, et al. Coronary
vasospasm and atrial fibrillation associated with
ondansetron therapy. Ann Pharmacother. 2009;43:532536.
Lombardi D, Crivellari D, Scuderi C, et al. Long-term,
weekly one-hour infusion of paclitaxel in patients with
metastatic breast cancer: a phase II monoinstitutional
study. Tumori. 2004;90:285-288.
Maister AH. Atrial fibrillation following physostigmine.
Can Anaesth Soc J. 1983;30:419-421.
Murdock CJ, Kyles AE, Yeung-Lai-Wah JA, et al. Atrial
flutter in patients treated for atrial fibrillation with
propafenone. Am J Cardiol. 1990;66:755-757.
Patel AK, Skatrud JB, Thomsen JH. Cardiac arrhythmias
due to oral aminophylline in patients with chronic
obstructive pulmonary disease. Chest. 1981;80:661-665.
Richardson JP. Theophylline toxicity associated with the
administration of ciprofloxacin in a nursing home
patient. J Am Geriatr Soc. 1990;38:236-238.
Henderson A, Wright DM, Pond SM. Management of
theophylline overdose patients in the intensive care unit.
Anaesth Intensive Care. 1992;20:56-62.
Varriale P, Ramaprasad S. Aminophylline induced atrial
fibrillation. Pacing Clin Electrophysiol. 1993;16:1953-1955.
Chazan R, Karwat K, Tyminska K, et al. Cardiac
arrhythmias as a result of intravenous infusions of
theophylline in patients with airway obstruction. Int J
Clin Pharmacol Ther. 1995;33:170-175.
Okada S, Teramoto S, Matsuoka R. Recovery from
theophylline toxicity by continuous hemodialysis with
filtration. Ann Intern Med. 2000;133:922.
Falk RH, Knowlton AA, Manaker S. Verapamil-induced
atrial fibrillation. N Engl J Med. 1980;318:640-641.
Garratt C, Linker N, Griffith M, et al. Comparison of
adenosine and verapamil for termination of paroxysmal
junctional tachycardia. Am J Cardiol. 1989;64:1310-1316.
Black DM, Delmas PD, Eastell R, et al. Once-yearly
zoledronic acid for treatment of postmenopausal
osteoporosis. N Engl J Med. 2007;356:1809-1822.
Mukamal KJ, Tolstrup JS, Friberg J, et al. Alcohol
consumption and risk of atrial fibrillation in men and
women: the Copenhagen City Heart Study. Circulation.
2005;112:1736-1742.
482
SECTION VI • Drug-Induced Cardiovascular Diseases
547. Abrahamsen B, Eiken P, Brixen K. Atrial fibrillation in
fracture patients treated with oral bisphosphonates. J
Intern Med. 2009;265:581-592.
548. Grosso A, Douglas I, Hingorani A, et al. Oral
bisphosphonates and risk of atrial fibrillation and flutter
in women: a self-controlled case-series safety analysis.
PLoS One. 2009;4:e4720.
549. Bunch TJ, Anderson JL, May HT, et al. Relation of
bisphosphonate therapies and risk of developing atrial
fibrillation. Am J Cardiol. 2009;103:824-828.
550. Loke YK, Jeevanantham V, Singh S. Bisphosphonates and
atrial fibrillation: systematic review and meta-analysis.
Drug Saf. 2009;32:219-228.
551. Smeets JL, Allessie MA, Lammers WJ, et al. The
wavelength of the cardiac impulse and reentrant
arrhythmias in isolated rabbit atrium: the role of heart
rate, autonomic transmitters, temperature, and calcium.
Circ Res. 1986;58:96-108.
552. Rensma PL, Allessie MA, Lammers WJ, et al. The length
of the excitation wave and susceptibility to reentrant
arrhythmias in normal conscious dogs. Circ Res.
1988;62:395-410.
553. O’Nunain S, Garratt C, Paul V, et al. Effect of intravenous
adenosine on human atrial and ventricular
repolarisation. Cardiovasc Res. 1992;26:939-1043.
554. O’Nunain S, Jennison S, Bashir Y, et al. Effects of
adenosine on atrial repolarization in the transplanted
human heart. Am J Cardiol. 1993;71:248-51.
555. Hewitt RE, Lissina A, Green AE, et al The bisphosphonate
acute phase response: rapid and copious production of
proinflammatory cytokines by peripheral blood
gammadelta T cells in response to
aminobisphosphonates is inhibited by statins. Clin Exp
Immunol. 2005;139:101-111.
556. Aviles RJ, Martin DO, Apperson-Hansen C, et al.
Inflammation as a risk factor for atrial fibrillation.
Circulation. 2003;108:3006-3010.
557. Sørensen HT, Christensen S, Mehnert F, et al. Use of
bisphosphonates among women and risk of atrial
fibrillation and flutter: population based case-control
study. BMJ. 2008;336:813-816.
558. Wijffels MC, Dorland R, Allessie MA. Pharmacologic
cardioversion of chronic atrial fibrillation in the goat by
class IA, IC, and III drugs: a comparison between
hydroquinidine, cibenzoline, flecainide, and d-sotalol. J
Cardiovasc Electrophysiol. 1999;10:178-193.
559. Wijffels MC, Dorland R, Mast F, et al. Widening of the
excitable gap during pharmacological cardioversion of
atrial fibrillation in the goat: effects of cibenzoline,
hydroquinidine, flecainide, and d-sotalol. Circulation.
2000;102:260-267.
560. Schreiber DH, DeFreest MS. Paroxysmal atrial fibrillation
precipitated by amiodarone-induced thyrotoxicosis five
months after cessation of therapy. J Emerg Med.
2006;31:61-64.
561. Kurt IH, Yigit T, Karademir BM. Atrial fibrillation due to
late amiodarone-induced thyrotoxicosis. Clin Drug Invest.
2008;28:527-531.
562. Masoni A, Alboni P, Malacarne C, et al. Effects of
dobutamine on electrophysiological properties of the
specialized conduction system in man. J Electrocardiol.
1979;12:361-370.
563. Lin CI, Chuang IN, Cheng KK, et al. Arrhythmogenic
effects of theophylline in human atrial tissue. Int J
Cardiol. 1987;17:289-297.
564. Fuster V, Rydén LE, Cannom DS, et al. ACC/AHA/ESC
2006 guidelines for the management of patients with
565.
566.
567.
568.
569.
570.
571.
572.
573.
574.
575.
576.
577.
578.
579.
580.
581.
582.
583.
atrial fibrillation: executive summary: a report of the
American College of Cardiology/American Heart
Association Task Force on Practice Guidelines and the
European Society of Cardiology Committee for Practice
Guidelines (Writing Committee to Revise the 2001
Guidelines for the Management of Patients With Atrial
Fibrillation). Circulation. 2006;114:700-752.
Phillips BG, Gandhi AJ, Sanoski CA, et al. Comparison of
intravenous diltiazem and verapamil for the acute
treatment of atrial fibrillation and atrial flutter.
Pharmacotherapy. 1997;17:1238-1245.
Minton NA, Henry JA. Treatment of theophylline
overdose. Am J Emerg Med. 1996;14:606-612.
Blomstrom-Lundqvist C, Scheinman MM, Aliot EM, et al.
ACC/AHA/ESC guidelines for the management of
patients with supraventricular arrhythmias: executive
summary: a report of the American College of
Cardiology/American Heart Association Task Force on
Practice Guidelines and the European Society of
Cardiology Committee for Practice Guidelines (writing
committee to develop guidelines for the management of
patients with supraventricular arrhythmias). J Am Coll
Cardiol. 2003;42:1493-1531.
Josephson GW, Stine RJ. Caffeine intoxication: a case of
paroxysmal atrial tachycardia. JACEP. 1976;5:776-778.
Clark JE, Simon WA. Cardiac arrhythmias after
phenylpropanolamine ingestion. Drug Intell Clin Pharm.
1983;17:737-738.
Somani PN, Jhingran SG. A case of paroxysmal atrial
tachycardia with block due to digitalis overdosage. Indian
Heart J. 1965;17:85-90.
Storstein O, Rasmussen K. Digitalis and atrial tachycardia
with block. Br Heart J. 1974;36:171-176.
Rose MR, Glassman E, Spencer FC. Arrhythmias
following cardiac surgery: relation to serum digoxin
levels. Am Heart J. 1975;89:288-294.
Storstein O, Hansteen V, Hatle L, et al. Studies on digitalis:
XIII. A prospective study of 649 patients on maintenance
treatment with digitoxin. Am Heart J. 1977; 93:434-443.
Bramlet DA, From AH. Double tachycardias associated
with digitalis intoxication. Chest. 1980;78:324-326.
Goren C, Denes P. The role of Holter monitoring in
detecting digitalis-provoked arrhythmias. Chest.
1981;79:555-558.
Chowdhry IH, Hariman RJ, Gomes JA, et al. Transient
digitoxic double tachycardia. Chest. 1983;83:686-687.
Moorman JR. Digitalis toxicity at Duke Hospital, 19731984.South Med J. 1985;78:561-564.
Bigger JT Jr. Digitalis toxicity. J Clin Pharmacol.
1985;25:514-521.
Desantola JR, Marchlinski FE. Response of digoxin toxic
atrial tachycardia to digoxin-specific Fab fragments. Am J
Cardiol. 1986;58:1109-1110.
Miura T, Kojima R, Sugiura Y, et al. Effect of aging on the
incidence of digoxin toxicity. Ann Pharmacother.
2000;34:427-432.
Laaban JP, Iung B, Chauvet JP, et al. Cardiac arrhythmias
during the combined use of intravenous aminophylline
and terbutaline in status asthmaticus. Chest.
1988;94:496-502.
Levine JH, Michael JR, Guarnieri T. Multifocal atrial
tachycardia: a toxic effect of theophylline. Lancet.
1985;1(8419):12-14.
Marchlinski FE, Miller JM. Atrial arrhythmias
exacerbated by theophylline. Response to verapamil
and evidence for triggered activity in man. Chest.
1985;88:931-934.
CHAPTER 25 • Supraventricular Arrhythmias
584. Sessler CN, Cohen MD. Cardiac arrhythmias during
theophylline toxicity: a prospective continuous
electrocardiographic study. Chest. 1990;98:672-678.
585. Emerman CL, Devlin C, Connors AF. Risk of toxicity in
patients with elevated theophylline levels. Ann Emerg
Med. 1990;19:643-648.
586. Bittar G, Friedman HS. The arrhythmogenicity of
theophylline: a multivariate analysis of clinical
determinants. Chest. 1991;99:1415-1420.
587. Hoffman BF, Bigger JT. Digitalis and allied cardiac
glycosides. In: Gilman AG, Goodman LS, Gilman A, eds.
Goodman and Gilman’s The Pharmacological basis of
Therapeutics. 6th ed. New York, NY: MacMillan
Publishing Co Inc;1985:729-760.
588. Cockcroft DW, Gault MH. Prediction of creatinine
clearance from serum creatinine. Nephron. 1976;16:31-41.
589. Schentag JJ, Bang AJ, Kozinski-Tober JL. Digoxin. In:
Burton ME, Shaw LM, Schentag JJ, et al, eds. Applied
Pharmacokinetics and Pharmacodynamics. 4th ed.
Philadelphia, PA: Lippincott Williams & Wilkins;
2005:410-439.
590. Markowitz SM, Stein KM, Mittal S, et al. Differential
effects of adenosine on focal and macroreentrant atrial
tachycardia. J Cardiovasc Electrophysiol. 1999;10:489-502.
591. Arsura EL, Solar M, Lefkin AS, et al. Metoprolol in the
treatment of multifocal atrial tachycardia. Crit Care Med.
1987;15:591-594.
592. Arsura E, Lefkin AS, Scher DL, et al. A randomized,
double-blind, placebo-controlled study of verapamil and
metoprolol in treatment of multifocal atrial tachycardia.
Am J Med. 1988;85(4):519-524.
593. Iseri LT, Fairshter RD, Hardemann JL, et al. Magnesium
and potassium therapy in multifocal atrial tachycardia.
Am Heart J. 1985;110:789-794.
594. Cohen L, Kitzes R, Shnaider H. Multifocal atrial
tachycardia responsive to parenteral magnesium. Magnes
Res. 1988;1:239-242.
595. McCord JK, Borzak S, Davis T, et al. Usefulness of
intravenous magnesium for multifocal atrial tachycardia
in patients with chronic obstructive pulmonary disease.
Am J Cardiol. 1998;81:91-93.
596. Eidelman DH, Sami MH, McGregor M, et al.
Combination of theophylline and salbutamol for
arrhythmias in severe COPD. Chest. 1987;91:808-812.
597. Rotstein Z, Battler A, Rath S, et al. Acute hemodynamic
and arrhythmogenic effects of high-dose intravenous
salbutamol in patients with chronic left ventricular
dysfunction. Clin Cardiol. 1990;13:468-472.
598. Franklin WH, Dietrich AM, Hickey RW, et al. Anomalous
left coronary artery masquerading as infantile
bronchiolitis. Pediatr Emerg Care. 1992;8:338-341.
599. Lin RY, Smith AJ, Hergenroeder P. High serum albuterol
levels and tachycardia in adult asthmatics treated with
high-dose continuously aerosolized albuterol. Chest.
1993;103:221-225.
600. Cook P, Scarfone RJ, Cook RT. Adenosine in the
termination of albuterol-induced supraventricular
tachycardia. Ann Emerg Med. 1994;24:316-319.
601. Keller KA, Bhisitkul DM. Supraventricular tachycardia: a
complication of nebulized albuterol. Pediatr Emerg Care.
1995;11:98-99.
602. Duane M, Chandran L, Morelli PJ. Recurrent
supraventricular tachycardia as a complication of
nebulized albuterol treatment. Clin Pediatr. 2000;39:673677.
603. Habashy D, Lam LT, Browne GJ. The administration of
beta2-agonists for paediatric asthma and its adverse
604.
605.
606.
607.
608.
609.
610.
611.
612.
613.
614.
615.
616.
617.
618.
619.
620.
621.
483
reaction in Australian and New Zealand emergency
departments: a cross-sectional survey. Eur J Emerg Med.
2003;10:219-224.
Trachsel D, Newth CJ, Hammer J. Adenosine for
salbutamol-induced supraventricular tachycardia.
Intensive Care Med. 2007;33:1676.
Zimmerman PM, Pulliam J, Schwengels J, et al. Caffeine
intoxication: a near fatality. Ann Emerg Med.
1985;14:1227-1229.
Price KR, Fligner DJ. Treatment of caffeine toxicity with
esmolol. Ann Emerg Med. 1990;19:44-46.
Beale MD, Pritchett JT, Kellner CH. Supraventricular
tachycardia in a patient receiving ECT, clozapine, and
caffeine. Convuls Ther. 1994;10:228-231.
Takayanagi K, Yamaguchi H, Hayashi T, et al.
Carbamazepine-induced bradycardia-tachycardia
syndrome with pharmacological analysis and concurrent
ECG monitoring. J Electrocardiol. 1990;23:85-88.
Kupchik M, Spivak B, Mester R, et al. Combined
electroconvulsive-clozapine therapy. Clin Neuropharmacol.
2000;23:14-16.
Caldicott LD, Hawley K, Heppell R, et al. Intravenous
enoximone or dobutamine for severe heart failure after
acute myocardial infarction: a randomized double-blind
trial. Eur Heart J. 1993;14:696-700.
Picano E, Mathias W Jr, Pingitore A, et al. Safety and
tolerability of dobutamine-atropine stress
echocardiography: a prospective, multicentre study: Echo
Dobutamine International Cooperative Study Group.
Lancet .1994;344:1190-1192.
Weissman NJ, Rose GA, Foster GP, et al. Effects of
prolonging peak dobutamine dose during stress
echocardiography. J Am Coll Cardiol. 1997;29:526-530.
Hennessy TG, Codd MB, Kane G, et al. Safety of
dobutamine stress echocardiography in 474 consecutive
studies. Coron Artery Dis. 1997;8:175-178.
Secknus MA, Marwick TH. Evolution of dobutamine
echocardiography protocols and indications: safety and
side effects in 3,011 studies over 5 years. J Am Coll
Cardiol. 1997;29:1234-1240.
Elhendy A, Geleijnse ML, van Domburg RT, et al. Gender
differences in the accuracy of dobutamine stress
echocardiography for the diagnosis of coronary artery
disease. Am J Cardiol. 1997;80:1414-1418.
Elhendy A, Valkema R, van Domburg RT, et al. Safety of
dobutamine-atropine stress myocardial perfusion
scintigraphy. J Nucl Med. 1998;39:1662-1666.
Elhendy A, van Domburg RT, Poldermans D, et al. Safety
and feasibility of dobutamine-atropine stress
echocardiography for the diagnosis of coronary artery
disease in diabetic patients unable to perform an exercise
stress test. Diabetes Care. 1998;21:1797-1802.
Elhendy A, van Domburg RT, Bax JJ, et al. Relation
between the extent of coronary artery disease and
tachyarrhythmias during dobutamine stress
echocardiography. Am J Cardiol. 1999;83:832-835.
Lu D, Greenberg MD, Little R, et al. Accelerated
dobutamine stress testing: safety and feasibility in
patients with known or suspected coronary artery
disease. Clin Cardiol. 2001;24:141-145.
Bountioukos M, Kertai MD, Schinkel AF, et al. Safety of
dobutamine stress echocardiography in patients with
aortic stenosis. J Heart Valve Dis. 2003;12:441-446.
Timperley J, Mitchell AR, Thibault H, et al. Safety of
contrast dobutamine stress echocardiography: a single
center experience. J Am Soc Echocardiogr. 2003;18:63167.
484
SECTION VI • Drug-Induced Cardiovascular Diseases
622. Tsutsui JM, Elhendy A, Xie F, et al. Safety of dobutamine
stress real-time myocardial contrast echocardiography. J
Am Coll Cardiol. 2005;45:1235-1242.
623. Kane GC, Hepinstall MJ, Kidd GM, et al. Safety of stress
echocardiography supervised by registered nurses: results
of a 2-year audit of 15,404 patients. J Am Soc Echocardiogr.
2008;21:337-341.
624. Gardner SF, Rutherford WF, Munger MA, et al. Druginduced supraventricular tachycardia: a case report of
fluoxetine. Ann Emerg Med. 1991;20:194-197.
625. Allhof T, Bender S, Banger M, et al. Atrial arrhythmia in a
woman treated with fluoxetine: is there a causal
relationship? Ann Emerg Med. 2001;37:116-117.
626. Wilson NJ, Adderley RJ, McEniery JA. Supraventricular
tachycardia associated with continuous furosemide
infusion. Can J Anaesth. 1991;38:502-505.
627. O’Driscoll BR. Supraventricular tachycardia caused by
nebulised ipratropium bromide. Thorax. 1989;44:312.
628. Anthonisen NR, Connett JE, Enright PL, et al.
Hospitalizations and mortality in the Lung Health Study.
Am J Respir Crit Care Med. 2002;166:333-339.
629. Smith RS, Warren DJ. Effects of high-dose intravenous
methylprednisolone on circulation in humans.
Transplantation. 1983;35:349-351.
630. Kumari R, Uppal SS. First report of supraventricular
tachycardia after intravenous pulse methylprednisolone
therapy, with a brief review of the literature. Rheumatol
Int. 2005;26:70-73.
631. Conway EE Jr, Walsh CA, Palomba AL. Supraventricular
tachycardia following the administration of
phenylpropanolamine in an infant. Pediatr Emerg Care.
1989;5:173-174.
632. Loughnan PM, McNamara JM. Paroxysmal
supraventricular tachycardia during theophylline therapy
in a premature infant. J Pediatr. 1978;92:1016-1018.
633. Ganz LI, Friedman PL. Supraventricular tachycardia. N
Engl J Med. 1995;332:162-173.
634. Ferguson JD, DiMarco JP. Contemporary management of
paroxysmal supraventricular tachycardia. Circulation.
2003;107:1096-1099.
Download