18
Drug-Induced QT-Interval Prolongation: Considerations for Clinicians Edward C. Li, Pharm.D., John S. Esterly, Pharm.D., Shaunte Pohl, Pharm.D., Shane D. Scott, Pharm.D., and Brian F. McBride, Pharm.D. Drug-induced proarrhythmia is a frequently encountered clinical problem and a leading cause for withdrawal or relabeling of prescription drugs. Suppression of the rapid component of the delayed rectifier potassium current, I Kr , represents the principal pharmacodynamic mechanism leading to heterogeneous prolongation of the ventricular action potential and prolongation of the QT interval clinically. However, the risk of proarrhythmia by QT-interval–prolonging drugs is variable and critically dependent on several factors leading to multiple reductions in the cardiac repolarization reserve. As antiarrhythmic drugs that prolong the QT interval are usually aggressively managed with continuous electrocardiogram monitoring and screening for drug interactions when administered to patients who have a high risk of sudden cardiac death, their risk of mortality is not increased. However, noncardiovascular QT-interval–prolonging drugs, which often produce less QT-interval prolongation compared with antiarrhythmic drugs, are found to be associated with increased rates of death in patients who have a markedly lower de novo risk of sudden cardiac death. Thus, it is important for clinicians, particularly pharmacists, to be cognizant of the levels of risk associated with varying degrees of QT-interval prolongation caused by drugs so that they can develop strategies to either prevent or reduce the risk of proarrhythmias. Key Words: antiarrhythmic agents, QT interval, QT prolongation, arrhythmia, atrial fibrillation, sudden cardiac death, torsade de pointes, pharmacogenomics. (Pharmacotherapy 2010;30(7):684–701) OUTLINE Physiology of Cardiac Conduction Reduced Repolarization Reserve Monophasic Action Potential Ventricular Repolarization Measurement and Heart Rate Correction of the QT Interval Guidelines for QT or QTc Interval Prolongation: How Much Is Too Much? Cardiovascular Drugs Antiarrhythmic Drugs That Primarily Suppress I Kr Multimechanistic Antiarrhythmics Prophylactic Strategies Drugs for Mental Health Disorders Methadone First-Generation Antipsychotics Second-Generation Antipsychotics Antimicrobial Agents Macrolides Fluoroquinolones Azole Antifungals Antineoplastic Agents Tyrosine Kinase Inhibitors Arsenic Trioxide Supportive and Emerging Agents Conclusion Arrhythmogenic syncope after starting quinidine therapy has been recognized for nearly a century. In the early 1960s, a case series was published that described patients with atrial fibrillation who, after quinidine-induced conversion to normal sinus rhythm, developed a drug-induced

Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

Embed Size (px)

Citation preview

Page 1: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

Drug-Induced QT-Interval Prolongation:Considerations for Clinicians

Edward C. Li, Pharm.D., John S. Esterly, Pharm.D., Shaunte Pohl, Pharm.D.,Shane D. Scott, Pharm.D., and Brian F. McBride, Pharm.D.

Drug-induced proarrhythmia is a frequently encountered clinical problem anda leading cause for withdrawal or relabeling of prescription drugs.Suppression of the rapid component of the delayed rectifier potassiumcurrent, IKr, represents the principal pharmacodynamic mechanism leading toheterogeneous prolongation of the ventricular action potential andprolongation of the QT interval clinically. However, the risk of proarrhythmiaby QT-interval–prolonging drugs is variable and critically dependent onseveral factors leading to multiple reductions in the cardiac repolarizationreserve. As antiarrhythmic drugs that prolong the QT interval are usuallyaggressively managed with continuous electrocardiogram monitoring andscreening for drug interactions when administered to patients who have ahigh risk of sudden cardiac death, their risk of mortality is not increased.However, noncardiovascular QT-interval–prolonging drugs, which oftenproduce less QT-interval prolongation compared with antiarrhythmic drugs,are found to be associated with increased rates of death in patients who have amarkedly lower de novo risk of sudden cardiac death. Thus, it is importantfor clinicians, particularly pharmacists, to be cognizant of the levels of riskassociated with varying degrees of QT-interval prolongation caused by drugsso that they can develop strategies to either prevent or reduce the risk ofproarrhythmias.Key Words: antiarrhythmic agents, QT interval, QT prolongation, arrhythmia,atrial fibrillation, sudden cardiac death, torsade de pointes, pharmacogenomics.(Pharmacotherapy 2010;30(7):684–701)

OUTLINE

Physiology of Cardiac ConductionReduced Repolarization ReserveMonophasic Action PotentialVentricular Repolarization

Measurement and Heart Rate Correction of the QTInterval

Guidelines for QT or QTc Interval Prolongation:How Much Is Too Much?

Cardiovascular DrugsAntiarrhythmic Drugs That Primarily Suppress IKr

Multimechanistic AntiarrhythmicsProphylactic Strategies

Drugs for Mental Health DisordersMethadoneFirst-Generation Antipsychotics

Second-Generation AntipsychoticsAntimicrobial Agents

MacrolidesFluoroquinolonesAzole Antifungals

Antineoplastic AgentsTyrosine Kinase InhibitorsArsenic TrioxideSupportive and Emerging Agents

Conclusion

Arrhythmogenic syncope after starting quinidinetherapy has been recognized for nearly a century.In the early 1960s, a case series was publishedthat described patients with atrial fibrillationwho, after quinidine-induced conversion tonormal sinus rhythm, developed a drug-induced

Page 2: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

proarrhythmia characterized as a pause-dependent polymorphic ventricular tachycardia.1

In 1966, this rhythm was also identified byFrançois Dessertenne as torsade de pointes (TdP;literally translated as “twisting of the points”[Figure 1]) because of the alternating axis of theQRS complex during the arrhythmia.2 Since thattime, drug-induced proarrhythmia has evolvedinto a common cause of hospital admissions, hasresulted in withdrawals of prescription drugsfrom the market by the government, and hasbecome a significant barrier to new drugdevelopment.3, 4

Virtually all drugs that cause TdP do so byincreasing the time for electrical recovery ofmyocardial tissue (i.e., repolarization) betweenheartbeats. Most, but not all, drug-inducedincreases in repolarization are the result ofsuppression of the rapid component of thedelayed rectifier potassium current (IKr).

Suppression of IKr often leads to prolongation ofthe heart rate–corrected QT (QTc) interval on theclinical electrocardiogram (ECG).5–8 Despite thepresence of risk factors leading to exaggeratedQT-interval prolongation, the risk of drug-induced TdP is imprecise and remains highlystochastic even among patients with the samerisk profile and equivalent QT intervals.9–15

These risk factors for TdP are as follows:

• Hypokalemia• Hypomagnesemia• Bradycardia• Bundle branch block• Ion channel polymorphisms (e.g., KCNH2,KCNQ1, SCNA5A)

• Female sex• Cardiovascular disease (chronic heart failure,

previous myocardial infarction)• Conversion from atrial fibrillation to normal

sinus rhythm• Rapid infusion or overdose of QTc-interval–

prolonging drugs• Prolonged QT interval at baseline• Family history of long QT interval

Many QT-interval–prolonging antiarrhythmicdrugs are administered in the hospital wherepatients receive continuous ECG monitoring.Rigorous patient selection, ECG monitoring, andscreening for drug interactions contribute to thelow risk of TdP and lack of increased mortality inpatients receiving most QT-prolongingantiarrhythmic drugs.9, 16–20 However, registrydata suggest that noncardiovascular QT-prolonging drugs, which often produce less QT-

685

From the National Comprehensive Cancer Network, FortWashington, Pennsylvania (Dr. Li); the Department ofPharmacy, Northwestern Memorial Hospital, Chicago,Illinois (Dr. Esterly); the Department of Pharmacy Practice,Midwestern University Chicago College of Pharmacy,Downers Grove, Illinois (Drs. Esterly and Pohl); theDepartments of Pharmacy and Psychiatry, North ChicagoVeterans Affairs Medical Center; Chicago, Illinois (Dr.Pohl); Baxter Healthcare, Deerfield, Illinois (Dr. Scott); andthe Marcella Niehoff School of Nursing and the StritchSchool of Medicine, Loyola University, Chicago, Illinois (Dr.McBride).

For reprints, visit http://www.atypon-link.com/PPI/loi/phco.For questions or comments, contact Brian F. McBride,Pharm.D., Marcella Niehoff School of Nursing, LoyolaUniversity Chicago, Building 102-4601, 2160 South FirstAvenue, Maywood, IL 60153; e-mail: [email protected].

Figure 1. Electrocardiogram of a patient with drug-induced QTc-interval prolongation and torsade de pointes.

Page 3: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

interval prolongation compared with antiarrhythmicdrugs, are associated with 2–3-fold increases inthe risk of sudden cardiac death.21, 22 The increasedrisk attributed to noncardiovascular QT-prolonging drugs is thought to be due to theconvergence of four main factors: clinical trialsthat are not designed to predict risk of TdP; atarget population that is large enough to producea significant proportion of cases of TdP despitelow estimates from clinical trials; pharmacokineticand pharmaco-dynamic drug interactions thatlead to an increased exposure of the drug; andprescription of these drugs by noncardiologistswho are less likely to be familiar with thepotential TdP risk conferred by them.23–26 Takentogether, these factors place an increasedimportance on the role of the clinical pharmacistin the prevention of drug-induced TdP. Pharmacistsmust understand the physiology of the cardiacconduction system and the degrees of acceptabledrug-induced QT-interval prolongation; theyshould also be aware of strategies for managingpatients who are receiving QT-prolonging drugs.

Physiology of Cardiac Conduction

Drug-induced QT-interval prolongationoriginates with either prolonged depolarization,reduced repolarization, or a combination of both.Prolonged depolarization is accomplished byadministering a drug that activates the latecomponent of the cardiac sodium current (INa).Reduced repolarization is accomplished throughsuppression of IKr or the simultaneoussuppression of IKr and the slowly activatingcomponent of the delayed rectifier potassiumcurrent (IKs).27–29 To our knowledge, there are nodrugs in clinical use that selectively suppress IKs.

Increases in late INa and/or reductions in IKrand IKs after drug block do not always predict thedegree of QT-interval prolongation. Indeed, thedegree of QT prolongation after exposure to aQT-prolonging drug is critically dependent onthe integrity of the redundant systems involvedin cardiac repolarization, such as IKs and post-translational modification of proteins involved inrepolarization.30 This redundancy is labeled“repolarization reserve” and considers the abilityof a drug to induce QT prolongation and/or TdPto be strongly dependent on the concomitantpresence of multiple reductions to the repolari-zation system including but not limited to thefollowing risk factors: hypokalemia, hypomagne-semia, female sex, bradycardia (or drugs slowingatrioventricular nodal conduction), and subclinical

mutations or polymorphisms in genes encodingcardiac ion channels that carry ion currents(mentioned above). However, the geneticgovernance of drug-induced QT prolongationand/or TdP remains unpredictable.5, 11–14, 31–32

Each of these risk factors either suppressesrepolarization directly or enhances drug-inducedsuppression of repolarization.

Although drugs and risk factors leading to QT-interval prolongation in patients decreaserepolarization reserve, decreases in repolarizationreserve are not the sole determinant forexaggerated QT prolongation and/or TdP.Multiple hypotheses regarding the genesis ofdrug-induced TdP have been proposed; however,one of the most well-described hypothesesincludes the convergence of three key electro-physiologic factors: decreased repolarizationreserve (i.e., QT-interval prolongation), increasedtransmural dispersion of repolarization, and theability to induce ectopic (i.e., premature)heartbeats caused by early afterdepolarizations.

Reduced Repolarization Reserve

With respect to reduced repolarization reserve,drug block of IKr and IKs and activation of late INado not represent the sole determinants of QT-interval prolongation. Indeed, an analysis of 249case reports of drug-induced TdP revealed thatthe convergence of multiple risk factors increasedthe risk for TdP.33 Serum potassium andmagnesium levels in the low or low-to-normalrange (potassium < 4 mEq/L and/or magnesium< 2 mg/dl) intensify IKr drug block and arepresent in approximately 40% of TdP cases.33–35

In addition, it was recently shown thatintracellular levels of these electrolytes correlatewith the duration of the QTc interval, thatpatients receiving diuretic therapy can haveintracellular electrolyte depletion despite normalserum levels, and that electrolyte repletion leadsto a shortening of the QTc interval.36, 37

Several studies have shown that compared withmale patients, female patients have dispropor-tionately longer QT intervals de novo and afterreceiving a QT-interval–prolonging drug and thatthis risk may be related to postpubescent sexhormones and is greatest during ovulation andmenstruation.12, 38–41 These factors may explainwhy 71% of all cases of drug-induced TdP occurin females.33

Bradycardia (heart rate < 60 beats/min) ordrugs slowing atrioventricular nodal conduction(e.g., �-blockers, digoxin, diltiazem, and

686

Page 4: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

verapamil) intensify drug block of potassiumchannels. This property is known as reverse usedependence and often occurs with drugs thatselectively suppress IKr.10

Finally, the importance of screening for drug-drug interactions cannot be understated. ManyQT-interval–prolonging drugs are substrates ofthe cytochrome P450 (CYP) enzyme system andwere identified in 39% of drug-induced TdPcases. The combination of multiple drugsblocking IKr or inhibition of the metabolism of IKrcan lead to high concentrations of the QT-prolonging drug, further reducing repolarizationreserve.33

Monophasic Action Potential

In the ventricular tissue, the QRS and QTintervals on the surface ECG approximate theventricular monophasic action potential (Figure 2).Each of the five sections (known as phases 0–4)of the monophasic action potential is producedby the synchronous action of cardiac ion channels,which transport ions across an electrochemicalgradient across the myocyte cell membrane.42

The balance of inward current and outwardcurrent activation, inactivation, and deactivationchanges as the cell undergoes depolarization andrepolarization; creating the monophasic actionpotential and thus allowing for the spontaneousbeating of myocytes.

When a selective IKr blocker such as ibutilideor dofetilide is used, these agents prolong theQTc by preferentially extending phase 3 of themonophasic action potential. This changes theshape of the action potential from the “spike anddome” morphology shown in Figure 2 to a moretriangular shape, referred to as “triangulation.”When triangulation occurs, the duration of timefor the monophasic action potential to reach 90%recovery from 40% recovery is prolonged,extending the vulnerable period where themyocytes are more susceptible to early after-depolarizations. Multimechanistic drugs, such asamiodarone, prolong the QTc by extending bothphases 2 and 3 of the monophasic actionpotential, preventing triangulation. Thisrepresents one possible explanation for whyamiodarone is associated with a markedly lowerrate of TdP compared with selective IKr blockerssuch as dofetilide, ibutilide, and sotalol.43

Ventricular Repolarization

Depolarization and repolarization of themyocardium act as electrical wavefronts, the

propagation of which may be visualized as thecrashing of two successive waves upon acoastline. The depolarization proceeds as awavefront, from the base of the ventricle, throughthe bundle of His and down the Purkinje fiberstoward the apex of the ventricle and outwardfrom the endocardium, through the midmyo-cardial layer and the epicardium. The repolar-ization wavefront, represented clinically by theQT interval, immediately succeeds the depolar-ization wavefront, following the same base toapex and endocardial to epicardial patterns.44

The QT-interval–prolonging drugs increase theduration of the repolarization wavefront andproduce a T wave that is not only prolonged buthas a reduction in the amplitude (i.e., T-waveflattening).

As the depolarization and repolarizationwavefronts progress through the myocardium,the monophasic action potential also exhibits asmall amount of heterogeneity in depolarizationand repolarization. This heterogeneity is

687

Figure 2. Synchronous activation and deactivation ofcardiac ion channels produce a monophasic actionpotential, which is represented clinically as the QRScomplex and T wave from which the QT interval is derived.

Page 5: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

commonly known as dispersion and exists in twoforms: temporal dispersion and spatialdispersion. Temporal dispersion is the variabilityin the velocity of the depolarization wavefront.At myocardial site A, depolarization begins at atheoretical time of 0 msec. At myocardial site B,depolarization begins 30 msec later. Althoughthe duration of the monophasic action potentialis the same, the two sites are activated at differenttimes. This difference in activation can create azone of myocardium that facilitates reentrantarrhythmias.28

Dispersion in repolarization, however, isaffected by the variability in basal to apicalrepolarization, as well as variability in transmuralrepolarization. Differences in basal to apicalrepolarization are measured as the differencebetween the minimum and maximum QTintervals on a 12-lead ECG, reported as QTdispersion; this is not considered a risk factor fordrug-induced TdP. The transmural dispersion ofrepolarization represents spatial heterogeneityacross the wall of the myocardium from theendocardial layer to the epicardial layer.28 This ismeasured from the peak of the T wave to the endof the T wave. Drug-induced increases in trans-mural dispersion of repolarization are consideredpart of a triad of arrhythmogenic propertiesassociated with the development of TdP.

Clinically used QT-interval–prolonging drugsthat increase spatial transmural dispersion ofrepolarization generally exhibit preferentialsuppression of IKr. Preferential suppression of IKrproduces a greater prolongation of the actionpotential in the midmyocardial layer of the hearttissue relative to the epicardial and endocardiallayers. One of the reasons that suppression of IKrincreases transmural dispersion of repolarizationis because the myocytes of the midmyocardium,known as M cells, have a reduced expression ofthe KCNQ1 potassium channel, which carries IKs.Reductions in IKs density in the M cells makerepolarization, and thus sensitivity to drug blockof the M cells, more dependent on IKr.28, 45

Therefore, pharmacologic suppression of IKr has agreater impact on M cells relative to epicardialand endocardial myocytes.

To illustrate the influence of transmuraldispersion of repolarization, consider the factthat amiodarone, dofetilide, and sotalol exertsimilar effects on the QT interval clinically.However, amiodarone is associated with amarkedly lower (< 2%) frequency of TdP.Because amiodarone exerts an effect on multipleinward and outward ion currents, it reduces

transmural dispersion of repolarization, makingthe prolongation of the action potential morehomogeneous.46–49 However, sotalol anddofetilide suppress only IKr, increase transmuraldispersion of repolarization, and are associatedwith a higher rate of TdP clinically. Exogenousand endogenous catecholamines can also increasetransmural dispersion of repolarization. Basicscience literature has identified a link between�2-adrenoreceptor stimulation, activation of IKs,and drug block of IKs.50 When IKs is activated inthe presence of drug block, the action potentialduration is paradoxically shortened in theendocardium and epicardium where IKs density ishigher.51, 52 This leads to a relatively longeraction potential duration in the M cells (whereIKs density is smaller) and thus increasedtransmural dispersion of repolarization.Therefore, the ability of a drug or combination ofdrugs to increase transmural dispersion ofrepolarization through suppression of IKr orenhancement of IKs should be considered as acomponent of the electrophysiologic principlesnecessary to develop drug-induced TdP becausein either instance, this inhomogeneity in theventricular action potential facilitates conductionof early afterdepolarizations.

The ability of a QT-interval–prolonging drug togenerate early afterdepolarizations represents thethird component of the torsadogenic triad. Earlyafterdepolarizations originate during phase 3 ofrepolarization, leading portions of the ventricularmyocardium to conduct the premature heartbeat.These activated myocytes will initiate a reentrantloop capable of stimulating the remainingventricular myocardium that failed to conduct theextra stimulus de novo.53–55 Thus, when theventricular myocardium experiences decreasedrepolarization reserve and increased dispersion ofrepolarization, the introduction of an earlyafterdepolarization can trigger a premature R waveemanating out of the preceding T wave (R-on-Tphenomenon) on the clinical ECG, with theensuing electrical reentry clinically observed asTdP.28 An emerging body of basic science evidencesuggests intramyocyte calcium overload as amechanism leading to the development of earlyafterdepolarizations and a role for the sodium-calcium exchange protein, NCX, which carries thesodium-calcium exchange current, INa-Ca.

The NCX protein can operate in two modes.In the forward mode, one calcium ion leaves thecell in exchange for three sodium ions enteringthe cell, creating a net inward current thatshortens the monophasic action potential. The

688

Page 6: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

reverse mode is where three sodium ions leavethe cell in exchange for one calcium ion enteringthe cell, which leads to intramyocyte calciumaccumulation, a prolongation of phase 3 of themonophasic action potential and therefore earlyafterdepolarizations. The NCX protein generallyoperates in reverse mode during myocardialischemia and heart failure due to an increasedintramyocyte sodium concentration.56 Multi-mechanistic antiarrhythmic drugs such asamiodarone and ranolazine inhibit NCX andsuppress early afterdepolarizations.29, 57 However,early afterdepolarization suppression bydronedarone is much less potent than that byamiodarone and thus permits genesis of TdP inanimal models.47, 58

Measurement and Heart Rate Correction of theQT Interval

Although guidelines for the clinical measure-ment of the QT interval are not yet adopted inthe United States, procedures used for theassessment of QT-interval prolongation in clinicaltrials can provide some direction.59 The reader isreferred to a detailed tutorial on the propermeasurement of the QT interval.59

The duration of the QT interval exhibits a greatdegree of variability with the heart rate. Therefore,correction factors are used to accurately predictdrug-induced changes in cardiac repolarization.This is particularly important since drugs thatreduce IKr and IKs exhibit reverse use dependence,a phenomenon in which their potency isincreased at slower heart rates. Although morethan a dozen correction formulas have beenproposed, the Bazett formula, QTc = QT/(RR)1/2,is the most commonly used in clinical practicedespite the fact that it overestimates the true QTinterval when the heart rate is greater than 60beats/minute.60 In 1992, a linear regressionformula developed from the Framingham HeartStudy, QTc = QT + 0.154(1 − RR), with the dataentered in seconds, was proposed and moreaccurately predicts drug-induced changes inrepolarization.60, 61 The disadvantage to clinicaluse of the Framingham correction formula,however, is that most drug-induced QTc prolon-gation is reported with the Bazett correctionformula. Thus, assessing the clinical risk ofdrug-induced TdP in patients approaching keycutoff points for QTc-interval prolongation (460,480, 500 msec) should include calculating theQTc by using both correction formulas, verifyingall interacting drugs (e.g., diuretics, �-blockers),

and identifying changes toward a more flattenedT-wave morphology and/or the presence of a Uwaves.27

The U wave is the sixth deflection observed onthe ECG, follows the T wave, and usually has thesame polarity as the T wave. In healthyindividuals, this is generally considered anincident finding as it usually represents an earlyafterdepolarization that did not result in apremature heart beat. In the context of drug-induced QT-interval prolongation, the presenceof the U wave (often referred to as the second Twave, T2) is concerning as it representsinterrupted repolarization resulting fromdiffering action potential duration in theepicardium, endocardium, and midmyocardiallayers.62 Further, the presence of U waves hasbeen associated with an increased frequency ofpremature ventricular contractions (an earlyafterdepolarization that conducts a prematureheartbeat) and is more common in patients whodevelop spontaneous ventricular tachycardiarelative to those patients who do not (39% vs8.7%, p<0.001).62–64 Therefore, it is generallyrecommended that U waves be included in themeasurement of the QT interval when QT-interval–prolonging drugs are administered.

The clinical trial guidelines, adopted by theEuropean Council for Proprietary and MedicinalProducts and under review by the U.S. Food andDrug Administration (FDA), recognize severalupper-limit values for drug-induced QTc-intervalprolongation. These values include absolute QTcintervals and changes in the QTc interval fromthe baseline (predrug) ECG. Absolute valuesinclude QTc intervals greater than 450, 480, and500 msec and changes from baseline greater than30 or 60 msec, with increasing upper-limit valuessignifying an increased risk for the developmentof TdP.51, 59 In fact, 92% of TdP cases occurredwhen the patient developed an antecedent QTcinterval greater than 500 msec.65–66 We thereforegenerally recommend that QTc-interval–prolonging drugs not be started in patients with abaseline QTc greater than 460 msec in theabsence of identifiable risk factors unlessprescribed by a cardiologist or if the patient is ona unit with 24-hour telemetry monitoring. Whenpatients reach the upper-limit values for QTcprolongation, the proarrhythmic risk associatedwith the regimen should be reevaluated andalternative treatment regimens considered.

Cardiovascular Drugs

Contrary to the notion that procedure-based

689

Page 7: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

therapy has supplanted antiarrhythmic pharmaco-therapy, both strategies are often used togetherfor two principal indications. The first isrestoration and maintenance of normal sinusrhythm in patients with a diagnosis of atrialfibrillation after electrical cardioversion or acatheter ablation procedure. The second is toreduce the frequency of device activation inpatients with an implantable cardioverterdefibrillator.16, 19, 67–79 In this section, we reviewantiarrhythmic drugs that predominantlysuppress IKr, multimechanistic antiarrhythmics,and prophylactic strategies to enhance rhythmcontrol and reduce the extent of QT-intervalprolongation and frequency of TdP.

Antiarrhythmic Drugs That Primarily Suppress Ikr

Ibutilide

One of the most effective agents for acutepharmacologic rhythm conversion, ibutilide alsomarkedly enhances the success rate of electricalcardioversion of atrial fibrillation or atrial fluttercompared with electrical cardioversion alone.80, 81

Ibutilide is administered as an 1-mg/50-mlinfusion over 10 minutes. A second 1-mginfusion may be administered if the patient hasnot converted to normal sinus rhythm within 20minutes of the start of the first infusion. As oneof the most potent antiarrhythmic compounds,ibutilide produces extensive QT-intervalprolongation, ranging from 60–100 msec inclinical trials, underscoring the need to monitorserum potassium and magnesium levels, inter-acting drugs, and heart rate after adminis-tration.82, 83 In addition, animal models indicate amarked increase in the development of earlyafterdepolarizations and transmural dispersion ofrepolarization.84, 85

Emerging evidence from the basic scienceliterature suggests that ibutilide is a substrate forthe drug efflux pump P-glycoprotein and thatpatients should be prescreened for use of drugsthat inhibit P-glycoprotein.86 In the absence ofclinical data for this interaction and out of anabundance of caution, we recommend discon-tinuation of P-glycoprotein inhibitors as soon aspossible before administration of ibutilide. Ifdiscontinuation is not possible, the clinicianshould be aware that the first dose of ibutilidemight be more likely to convert atrial fibrillation,exaggerate QT-interval prolongation, andpossibly increase the risk of TdP. The oneexception to this recommendation is concomitantadministration of amiodarone with ibutilide.

Early clinical trials of ibutilide enrolled patientswho were receiving concomitant long-termamiodarone therapy. Compared with patientswho did not receive amiodarone, no significantdifference was noted in the frequency of ven-tricular arrhythmias. This was due in large partto the multimechanistic nature of amiodarone. Forother compounds that inhibit P-glycoprotein,prophylactic administration of intravenousmagnesium may reduce the extent of QT-intervalprolongation. Taken together, it is not surprisingthat the 5% rate of TdP after ibutilide administra-tion is among the highest of any antiarrhythmicdrug.

Sotalol and Dofetilide

Sotalol is a racemic mixture in which thedextro-isomer suppresses IKr and the levo- isomeris a nonselective �-adrenoreceptor antagonist.On the other hand, dofetilide is the only pure IKrantagonist available for clinical use in the UnitedStates. For the treatment of atrial fibrillation,50–60% of patients receiving either sotalol ordofetilide remained in normal sinus rhythm after1 year. Either agent can be used safely in patientswith left ventricular dysfunction withoutaffecting survival.9, 70, 87–90

Sotalol and dofetilide have similar effects onthe QTc interval. Sotalol produces QTc-intervalincreases from baseline of 25, 40, and 50 msec inpatients receiving 80-, 120-, and 160-mg doses,respectively, which translates into an average4.3% risk of TdP.91 Furthermore, the �-adrenoreceptor antagonist properties of sotalolmay influence the occurrence of TdP byincreasing its reverse use–dependent properties.Dofetilide also has a linear dose–QTc-intervalrelationship of 25 msec for each ng/ml of plasmadofetilide concentration, which translates into a25–50-msec increase in QTc over the dosingrange in subjects free of renal impairment,decreases to 20–30 msec after 3 weeks of therapy,and an average 2% increase in TdP.92–94 Bothdrugs increase transmural dispersion ofrepolarization and induce early afterdepolar-izations at concentrations used in clinicalpractice.95, 96

Although the risk of TdP with sotalol isarguably higher than the estimates for dofetilide,the dosing protocol and restrictions on the latterare the result of the more stringent regulatoryenvironment after the withdrawal of thenoncardiovascular QT-interval–prolonging drugs,terfenadine and cisapride. Regardless, hospitalinitiation protocols and maintenance of sotalol

690

Page 8: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

therapy should be subject to the same restrictionsas dofetilide germane to prescriber access, druginteractions, electrolyte level management, andpostadministration QTc-interval prolongationmanagement.

Multimechanistic Antiarrhythmics

Amiodarone

Amiodarone, a multimechanistic ion channelantagonist and antiadrenergic compound,29, 97–99

is the most potent antiarrhythmic in terms of theproportion of patients remaining in normal sinusrhythm 12 months after cardioversion from atrialfibrillation and the reduction in ventriculartachyarrhythmia burden in patients who receivedan implantable cardioverter defibrillator. Thismarked clinical response to chronic therapy withamiodarone is the result of not only suppressionof a number of key ion currents, including INa,ICaL, INa-CaR, IKr, and IKs, but also downregulationof the ion channel proteins that carry thesecurrents.100 Although amiodarone prolongs theQTc interval by 30–60 msec, it reverses trans-mural dispersion of repolarization and suppressesearly afterdepolarizations induced by sotalol inanimal models of TdP. These features confer thelowest TdP risk of any antiarrhythmic drug (<0.5%).46–49 Its noncardiovascular adverse-effectprofile spawned the development of several newantiarrhythmic compounds including azimilide,ranolazine, and dronedarone.

Dronedarone

Dronedarone, a deiodinated congener toamiodarone with a nearly identical electro-physiologic profile, recently demonstratedincreased mortality among patients with heartfailure and the inability to suppress earlyafterdepolarizations, thus limiting the use of thisdrug despite a marked reduction in noncardio-vascular toxicity.47, 101, 102

Ranolazine

Ranolazine is used as a second-line adjunctivetherapy for chronic stable angina when standardof care with �-adrenergic antagonists, calciumchannel antagonists, and/or long-acting nitratesbecome suboptimal or fail. Initial use ofranolazine was limited because of concernsregarding QTc-interval prolongation of 6–12msec with FDA-approved dosages of 500–1000mg twice/day. During clinical development, amore complete electrophysiologic evaluation of

ranolazine was performed. A series ofelectrophysiology experiments using cellular andanimal models indicate that ranolazine preventedsotalol-induced transmural dispersion ofrepolarization, early afterdepolarizations, andsuppressed TdP through antagonism of INa, IKr,IKs, and activation of calcium efflux by NCXsimilar to amiodarone.29, 103–105 These data werefurther supported by data from the MetabolicEfficiency with Ranolazine for Less Ischemia inNon–ST-Elevation Acute Coronary Syndromes–Thrombolysis in Myocardial Infarction(MERLIN-TIMI) 36 trial that demonstrated thatpatients with an acute coronary syndrome eventand treated with ranolazine 2000 mg/day in anintravenous to oral hybrid regimen had signifi-cantly fewer episodes of ventricular tachycardia,supraventricular tachycardia, or new-onset atrialfibrillation.106 Furthermore, we found nopublished cases of TdP induced by sustained-release ranolazine in the 10 years since the drugbegan clinical development. These data have ledthe manufacturer to proceed with clinicaldevelopment of ranolazine as a potentialantiarrhythmic compound.

Prophylactic Strategies

Mexiletine

For more than 2 decades, electrophysiologistshave used mexiletine and/or magnesium inconjunction with QT-interval–prolongingantiarrhythmic drugs to improve control ofventricular arrhythmias and reduce the risk ofventricular proarrhythmia in patients receivingan IKr blocker.107, 108 Mexiletine is an orallyavailable lidocaine analog and suppresses INa.When coadministered with potassium channelantagonists such as quinidine or sotalol,mexiletine shortens the QT interval, reduces thefrequency of TdP, and improves control ofventricular arrhythmias relative to quinidine orsotalol alone.107 Animal models also indicate thatmexiletine reduces transmural dispersion ofrepolarization and early afterdepolarizations afterexposure to sotalol.52, 108, 109 Adverse effects withmexiletine were limited to nausea. Mexiletineshould be used in patients who have break-through arrhythmias while taking a maximallytolerated dose of dofetilide, sotalol, or amiodarone.

Magnesium

In one study, oral magnesium L-lactate 162 mgadministered with either sotalol or dofetilide to

691

Page 9: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

patients with implantable cardioverter defibril-lators shortened their QTc intervals, especially inthose receiving loop diuretics for advanced heartfailure.36 The strategy was associated with nomagnesium-related adverse effects except for onecase of diarrhea. No reduction in TdP frequencywas reported in this 72-hour study, and the long-term effects of oral magnesium supplementationon the frequency of drug-induced TdP have notbeen evaluated. Since the influence of oralmagnesium supple-mentation on the efficacy ofdofetilide or sotalol has not been established, thepractice of magnesium repletion in patientsreceiving dofetilide or sotalol is uncommon andshould be used only when patients have apotassium level less than 4 mEq/L or amagnesium level less than 2 mg/dl.

For patients receiving ibutilide, the data forcoadministration of prophylactic magnesium area bit more convincing. When coadministeredwith ibutilide, magnesium 2 g over 10 minutesbefore the first ibutilide infusion and 2 g over 60minutes after the first ibutilide infusionprevented QTc-interval prolongation, suppressedearly afterdepolarizations, and facilitatedibutilide-induced cardioversion, particularly inpatients with impaired left ventricular functionfor whom the rate of TdP can be higher.110, 111

There were no magnesium-associated adverseevents in this study, and a 38% reduction wasnoted in the frequency of TdP (p=0.338).112

However, the study was not powered to detect adifference in the frequency of TdP.112 Given thisevidence, prophylactic magnesium is not oftenused to prevent TdP but to enhance the efficacyof ibutilide and reduce cost by decreasing theneed for direct current cardioversion.112

Prophylactic magnesium is not listed in theAmerican Heart Association guidelines foradvanced cardiac life support.113

Propafenone and Amiodarone

In clinical practice, it is common to useelectrical or pharmacologic cardioversion foracute restoration of sinus rhythm whileintroducing a second agent for the maintenanceof sinus rhythm. Two options for maintenancetherapy, propafenone and amiodarone, can helpreduce the risk of QT-interval prolongation andTdP associated with acute cardioversion.82, 83 Inpatients with structurally normal hearts (e.g., nocoronary artery disease or left ventricularhypertrophy), the ability of propafenone tosuppress INa shortens the ventricular action

potential, prevents ibutilide-induced QT-intervalprolongation, suppresses premature beats (i.e.,early afterdepolarizations), facilitates cardioversionto normal sinus rhythm, and has one of thehighest rates of sinus rhythm at 1 year.114–116 Forpatients with structural heart disease who receivecardioversion, the addition of amiodarone tomaintain sinus rhythm reduces the frequency ofTdP because of its effects on multiple ioncurrents, reductions in transmural dispersion ofrepolarization, and suppression of earlyafterdepolarizations.117

Drugs for Mental Health Disorders

Psychiatric disorders for which drug therapy isprescribed include substance abuse, majordepressive disorder, and schizophrenia. Each ofthese disorders is associated with an increasedrisk of cardiovascular disease and thus higherbaseline QT intervals relative to those in healthysubjects.118–127 Drugs used in the treatment ofmental health disorders that increase the QTinterval include methadone, as well as first- andsecond-generation antipsychotics.

Methadone

Methadone is a µ-opioid–receptor agonist usedfor maintenance therapy in opioid-dependentpatients (including illicit drug abusers), as wellas for treatment of moderate-to severe pain. TheS-isomer of methadone suppresses IKr amplitudeand prolongs the QTc interval by 14.1 msec at amean ± SD dose of 80 ± 32 mg, and at least 18cases of TdP associated with S-methadone havebeen reported in the literature.128, 129

An independent multidisciplinary expert panelon the cardiac effects of methadone recentlypublished guidelines for QT-interval screening inmethadone treatment.130 These guidelinesrecommend a thorough history and physicalexamination, a pretreatment ECG, and a secondECG 30 days after start of treatment or a changein dosage. The panel specifically recommendsincreased monitoring when a patient has a QTinterval greater than 450 msec. Discontinuationof methadone in patients with a QT intervalgreater than 500 msec is advised. Since poornutritional status is a common comorbid disorderin this population, aggressive monitoring andrepletion of serum potassium concentration to 4mEq/L or greater and magnesium to 2 mg/dl orgreater are highly recommended.

Although buprenorphine is the only FDA-

692

Page 10: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

approved alternative to methadone, diacetyl-morphine has been shown to be more efficaciousthan methadone for the treatment of drug addic-tion. Like buprenorphine, diacetylmorphine, toour knowledge, does not prolong the QTinterval.131, 132

First-Generation Antipsychotics

First-generation antipsychotics are recom-mended for use in patients with schizophreniawho had a partial response or no response to asecond-generation antipsychotic.133 Haloperidolis also used in the setting of acute agitation.134

Thioridazine, chlorpromazine, and haloperidolsuppress IKr, prolong the QT interval by 15–30msec at therapeutic doses (similar to long-termdofetilide therapy), and are associated withTdP.135–140 Although thioridazine causes thegreatest QTc-interval prolongation, chlorpromazineand haloperiodol have also been reported toproduce TdP, particularly when combined withother compounds that prolong the QTc intervalor inhibitors of the CYP2D6 enzyme, the principalmetabolic pathway for these compounds.141–145

When used for in-hospital agitation, intravenoushaloperidol produced a dose-dependent increasein the risk of a QTc greater than 500 msec andthe risk of TdP.146 Alternatives to the use ofhaloperidol in the setting of acute agitationinclude verbal deescalation of the patient,parenteral second-generation antipsychotics (e.g.,olanzapine) that do not prolong the QT interval,and use of benzodiazepines. If alternatives tohaloperidol are not effective, patients receivingintravenous or intramuscular haloperidol shouldreceive continuous ECG monitoring as soon asthey are stabilized.134 As with the treatment ofaddiction, the prevalence of poor nutritionalstatus as a comorbid disorder requires thatclinicians monitor and replete serum potassiumlevels to at least 4 mEq/L and magnesium to atleast 2 mg/dl.

Second-Generation Antipsychotics

Second-generation antipsychotics includeclozapine, quetiapine, olanzapine, risperidone,paliperidone, ziprasidone, and aripiprazole. Allof these drugs suppress IKr to varying degrees,with clozapine, olanzapine, aripiprazole, andpaliperidone being the least potent and devoid ofsignificant QTc prolongation clinically.135, 137,

147–149 Quetiapine increases the QTc interval in adose-dependent fashion by an average of 15–20msec in the presence of CYP3A4 inhibitors.137, 150

Torsade de pointes was reported in patientstreated with quetiapine and who had concomi-tant risk factors (e.g., bradycardia, hypomagne-semia) for TdP.151 Risperidone increases the QTcby 10 msec, can induce early afterdepolariza-tions, increases transmural dispersion of repolar-ization, and has also been reported to causeTdP.135, 152 Risperidone is also metabolized by theCYP2D6 enzyme.153, 154 Inherent genetic vari-ability in the CYP2D6 enzyme leads to genotype-dependent effects on the QTc interval. Patientshomozygous for the wild-type CYP2D6 enzymehad a significantly lower QTc (400 vs 418 msec,p<0.05) compared with those who wereheterozygous for the *4 variant (i.e., presentingwith one copy of the wild-type [*1] CYP2D6gene and one copy of the variant CYP2D6gene).153 Ziprasidone prolongs the QT interval ina dose-dependent fashion, but the effect plateausat 20 msec.135, 137 The reason for this plateau isthought to be the concomitant suppression of INaat increasing doses, which can mitigate the effectof IKr suppression on the QT interval and has notbeen reported to cause TdP.135

In patients requiring treatment with a second-generation antipsychotic, treatment witholanzapine, paliperidone, and aripiprazole carriesthe least risk of TdP. However, each second-generation antipsychotic has differing affinitiesfor dopamine receptor subtypes, which maynecessitate the use of a QTc-interval–prolongingsecond-generation antipsychotic. In the casewhere a QTc-prolonging second-generationantipsychotic must be used, the QT intervalshould be verified before starting the drug, at 7days after initiation, and after any changes indosing. Adequate screening and prevention ofrisk factors for QTc-interval prolongation shouldbe mitigated aggressively.

Antimicrobial Agents

Prolonged QTc interval and TdP have beenassociated with multiple antimicrobial compounds.Macrolides, fluoroquinolones, azole antifungals,and pentamidine represent the most commonlyimplicated compounds.155, 156 Antimicrobialagents less commonly implicated includetrimethoprim-sulfamethoxazole, clindamycin,and metronidazole.33, 157 Drug interactions are ofparamount concern since many of theseantimicrobials implicated in the development ofTdP suppress not only IKr but also substrates ofthe CYP enzyme system. Therefore, coadminis-tration of inhibitors of the CYP enzyme system

693

Page 11: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

(predominantly CYP3A4 and CYP2C9 inhibitors)should be avoided since inhibition of CYPenzyme systems could raise plasma concentra-tions of antimicrobials, leading to suppression ofIKr, prolongation of the QTc interval, anddevelopment of TdP.158

Macrolides

Erythromycin and clarithromycin possessintrinsic QTc-interval–prolonging effects bysuppressing IKr.159, 160 Several animal studiesindicate that erythromycin and clarithromycinincrease transmural dispersion of repolarizationand facilitate the development of early afterdepo-larizations.161, 162 Macrolides produce a “doublehit” on cardiac repolarization reserve since theysuppress IKr and inhibit their own metabolismthrough CYP3A4. A retrospective case analysisrevealed that erythromycin and clarithromycinwere responsible for 53% and 36% of the cases ofantibiotic-associated TdP, respectively.156, 163

Further, sudden cardiac death associated witherythromycin was increased 5-fold whenerythromycin was coadministered with a CYP3A4inhibitor (incidence rate ratio 5.35, 95%confidence interval 1.72–16.64, p=0.004).22, 164–166

Similar evaluations of clarithromycin are inprogress, given these data and case reports of TdPwith clarithromycin.26, 167–173

Fluoroquinolones

Fluoroquinolones are widely used antibioticagents because of their broad spectrum ofantimicrobial activity and relatively safe adverse-effect profile.174–176 Ciprofloxacin, levofloxacin,and moxifloxacin are associated with case reportsof TdP. Based on published data, moxifloxacin isassociated with the greatest risk of QTc-intervalprolongation of all of the fluoroquinolones.33, 177, 178

Therefore, moxifloxacin, out of an abundance ofcaution, should be reserved for treatment inthose patients who fail therapy with anotherfluoroquinolone.

Azole Antifungals

Azole antifungal agents exert their effectthrough the inhibition of 14-�-demethylase.This fungal CYP enzyme is responsible forergosterol synthesis and is homologous to severalhuman CYP enzymes. As a result, all of thecurrently available azoles—ketoconazole,fluconazole, itraconazole, voriconazole, andposaconazole—inhibit CYP3A4 to varying

degrees, with ketoconazole being the most potentCYP3A4 inhibitor and fluconazole being a potentCYP2C9 inhibitor. We found no evidencesuggesting that members of this class, other thanketoconazole, directly suppress IKr to an appre-ciable extent. Indeed, this group of compoundsindirectly contributes to TdP by increasing theplasma concentrations of CYP3A4 substratesknown to prolong the QTc interval.179–185

Therefore, in the setting of concomitant QT-interval–prolonging drugs that are also CYP3A4or P-glycoprotein substrates, the offendingCYP3A4 or P-glycoprotein substrates should beremoved or, if sensitivities of the fungal pathogenpermit, a switch to fluconazole should beconsidered.

Antineoplastic Agents

The conventional focus of adverse effects dueto cancer pharmacotherapy has been on mini-mizing or managing myelosuppression, emesis,anemia, alopecia, and other adverse effectsconsistent with traditional cytotoxic chemotherapy.However, innovations in mapping the humangenome over the past decade have allowed theparadigm of cancer pharmacotherapy to shiftfrom traditional “cytotoxic” chemotherapy agentstoward a molecular “targeted therapy” approachby using biologics and small molecule receptorinhibitors. These agents are associated with adifferent adverse-effect profile compared withthat of older antineoplastics because of differentpharmacologic properties. Preclinical studiessuggest that prolongation of the QT interval is acommon thread among the targeted therapyagents. Consequently, cardiac rhythm monitoringhas become increasingly important in clinicaltrials that use these novel agents. Furthermore,clinicians will need to be aware of these cardiacmonitoring recommendations as these drugstransition from investigational agents into themarketplace.186 The National Cancer Institute(NCI) Common Terminology Criteria for AdverseEvents (CTCAE) standardizes the severity ratingof adverse events, including QTc-intervalprolongation, into five possible grades for clinicaltrial purposes.187 By definition, grade 1 is a mildadverse event, whereas a grade 5 event causesdeath. Table 1 shows a comparison between theNCI CTCAE standard for QT prolongation andthe proposed FDA standard.

Tyrosine Kinase Inhibitors

Of the novel types of anticancer therapy,

694

Page 12: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

tyrosine kinase inhibitors are the most proliferative.Since 2001, seven tyrosine kinase inhibitors havebeen introduced into the marketplace for thetreatment of a variety of cancers (e.g., breast,lung, kidney, pancreatic, and leukemia). The riskof QT-interval prolongation is mostly of concernwith tyrosine kinase inhibitors that target multipletyrosine kinase receptors—nilotinib, dasatinib,and sunitinib. In vitro studies suggest that themechanism of QT prolongation is suppression ofIKr in a concentration-dependent manner.186 Therate of QT prolongation from clinical trials is low(< 3%),188–190 but the potential for life-threateningTdP warrants consideration and strict monitoringin some cases.

In clinical trials, sudden deaths attributed toQT-interval prolongation have been reportedwith nilotinib, and the frequency of TdP inpatients receiving sunitinib has been reported tobe less than 0.1%.189, 191 The maximum QT-interval prolongation by nilotinib was 18 msec,and a black-box warning about QT-intervalprolongation and sudden death is included in theFDA-approved package labeling.189 Dasatinib,nilotinib, and sunitinib are also CYP3A4 sub-strates, and a dosage reduction should beconsidered if administration with a concomitantCYP3A4 inhibitor is necessary. In addition, foodinteracts with nilotinib by increasing thebioavailability, and therefore nilotinib should betaken on an empty stomach. There are clearECG monitoring requirements when a patientbegins therapy with nilotinib. The frequency ofmonitoring and specific dosage adjustmentsbased on these measurements can be found in theproduct labeling.189

Arsenic Trioxide

Infamously known as a poison, arsenic trioxideis an inorganic compound approved by the FDAfor the treatment of relapsed or refractory acute

promyelocytic leukemia displaying translocationbetween chromosomes 15 and 17 or promyelocyticleukemia–retinoic acid receptor-� gene expression.192

Arsenic trioxide is associated with many adversereactions and can affect many organ systems; itsproduct labeling carries a black-box warning forECG abnormalities, including QT-intervalprolongation and TdP, although the mechanismof action is unclear.

The risk of QT-interval prolongation and TdPwith this agent is clinically significant. In aphase II clinical trial, arsenic trioxide caused aQT prolongation of more than 500 msec in 16(40%) of 40 patients, with one patient experi-encing TdP.193 A case report describes threepatients who experienced TdP while receivingarsenic trioxide for the treatment of acutemyeloid leukemia or myelodysplastic syndrome.194

The doses of arsenic trioxide ranged from 10–20mg/day, and all three patients had coexisting lowserum potassium and magnesium levels. Despiterigorous electrolyte replacement and otherinterventions (including cardioversion), all threepatients died.

The National Comprehensive Cancer Networkguidelines recommend the following for arsenictrioxide–induced QT-interval prolongation:ECG, serum electrolyte levels (potassium,magnesium, calcium), and serum creatinineconcentration should be assessed for abnor-malities before arsenic trioxide therapy isstarted.192 If the absolute value of the QT intervalrises above 500 msec during therapy, immediatecorrective action should be taken. Discontinu-ation of arsenic trioxide should be stronglyconsidered. Reinitiation may be considered onlywhen the QT interval decreases below 460 msecand all other abnormalities have been corrected.

Supportive and Emerging Agents

In addition to tyrosine kinase inhibitors and

695

Table 1. Standards for QTc-Interval Prolongation

Grade NCI CTCAE Standard187 Proposed FDA Standard59

1 (mild) QTc 450–470 msec QTc 450–479 msec or ≥ 30 msec above baseline

2 (moderate) QTc 470–499 msec or ≥ 60 msec above baseline QTc 480–499 msec or ≥ 60 msec above baseline

3 (severe) QTc > 500 msec QTc > 500 msec

4 (life-threatening QTc > 500 msec; life-threatening signs oror disabling) symptoms (e.g., arrhythmia, chronic heart

failure); torsade de pointes

5 (fatal) DeathQTc = heart rate–corrected QT; NCI = National Cancer Institute; CTCAE = Common Terminology Criteria for Adverse Events; FDA = U.S. Foodand Drug Administration.

Page 13: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

arsenic trioxide, other agents are listed as havingthe potential to cause QT-interval prolongation.Some data exist in the form of case reports and/orin vitro experiments for other antineoplastics andsupportive care agents such as anthracyclines,antiemetics (such as serotonin receptor antago-nists, droperidol, metoclopramide), octreotide,and tamoxifen, but there is generally littleconcern for developing QT prolongation and TdPin day-to-day practice with these agents.195–198

Recently, a black-box warning for QT prolon-gation and TdP was added to droperidol’sproduct labeling, but it has been debated whetherthis addition was necessary.199–201

New anticancer agents are likely to continue tobe evaluated for QT-interval prolongation.Histone deacetylase inhibitors such as vorinostatand romidepsin (depsipeptide) have been shownto prolong the QT interval in clinical studies.195,

196, 198–202 Baseline magnesium and potassiumlevels should be assessed before starting therapywith romidepsin, and ECG monitoring may beconsidered for those at high risk for developingQT-interval prolongation. In addition, heat-shock protein 90 inhibitors are in clinical trialsand will likely cause clinically significant QTprolongation through blocking the maturationand trafficking of KCNH2.203 Other classes in theoncology pipeline that may induce QT prolon-gation include vascular disruption agents,farnesyl protein transferase inhibitors, andprotein kinase C inhibitors.186

Conclusion

Drug-induced QT-interval prolongation repre-sents a key component of drug management forpharmacists in all areas of practice. Recognizingthe important levels of risk associated with varyingdegrees of QT prolongation conferred by drugscan prevent or reduce the risk of proarrhythmia.To properly assign a level of risk and provide anappropriate intervention to improve patient care,pharmacists need to assess baseline risk factorsfor QT prolongation and place a special emphasison the convergence of the intensity of drug-induced suppression of IKr, the ability of the drugto increase transmural dispersion of repolarization,and the ability of the offending drug to induceearly afterdepolarizations.

References1. Selzer A, Wray HW . Quinidine syncope: paroxysmal

ventricular fibrillation occurring during treatment of chronicatrial arrhythmias. Circulation 1964;30:17–26.

2. Dessertenne F. Ventricular tachycardia with 2 variable

opposing foci [in French]. Arch Mal Coeur Vaiss 1966;59:263–72.

3. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein DU,Wolfe SM, Bor DH. Timing of new black box warnings andwithdrawals for prescription medications. JAMA 2002;287:2215–20.

4. Wysowski DK, Swartz L. Adverse drug event surveillanceand drug withdrawals in the United States, 1969–2002: theimportance of reporting suspected reactions. Arch Intern Med2005;165:1363–9.

5. Kamiya K, Niwa R, Mitcheson JS, Sanguinetti MC .Molecular determinants of hERG channel block. MolPharmacol 2006;69:1709–16.

6. Mitcheson JS, Chen J, Lin M, Culberson C, Sanguinetti MC.A structural basis for drug-induced long QT syndrome. ProcNatl Acad Sci U S A 2000;97:12329–33.

7. Straus SMJM, Kors JA, De Bruin ML, et al. Prolonged QTcinterval and risk of sudden cardiac death in a population ofolder adults. J Am Col Cardiol 2006;47:362–7.

8. Algra A, Tijssen JG, Roelandt JR, Pool J, Lubsen J. QTcprolongation measured by standard 12-lead electrocardiographyis an independent risk factor for sudden death due to cardiacarrest. Circulation 1991;83:1888–94.

9. Torp-Pedersen C, Moller M, Bloch-Thomsen PE, et al.Dofetilide in patients with congestive heart failure and leftventricular dysfunction. N Engl J Med 1999;341:857–65.

10. Yang T, Roden DM. Extracellular potassium modulation ofdrug block of IKr: implications for torsade de pointes andreverse use-dependence. Circulation 1996;93:407–11.

11. Pham TV, Sosunov EA, Gainullin RZ, Danilo P Jr, Rosen MR.Impact of sex and gonadal steroids on prolongation ofventricular repolarization and arrhythmias induced by IK-blocking drugs. Circulation 2001;103:2207–12.

12. Drici MD, Burklow TR, Haridasse V, Glazer RI, Woosley RL.Sex hormones prolong the QT interval and downregulatepotassium channel expression in the rabbit heart. Circulation1996;94:1471–4.

13. Houltz B, Darpo B, Swedberg K, et al. Comparison of QTdispersion during atrial fibrillation and sinus rhythm in thesame patients, at normal and prolonged ventricularrepolarization. Europace 2000;2:20–31.

14. Napolitano C, Schwartz PJ, Brown AM, et al. Evidence for acardiac ion channel mutation underlying drug-induced QTprolongation and life-threatening arrhythmias. J CardiovascElectrophysiol 2000;11:691–6.

15. Goldberg R, Bengtson J, Chen Z, Anderson K, Locati E, LevyD. Duration of the QT interval and total and cardiovascularmortality in healthy persons (the Framingham heart studyexperience). Am J Cardiol 1991;67:55–8.

16. Connolly SJ, Dorian P, Roberts RS, et al. Comparison of �-blockers, amiodarone plus �-blockers, or sotalol forprevention of shocks from implantable cardioverterdefibrillators: the OPTIC study—a randomized trial. JAMA2006;295:165–71.

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

18. Wyse DG, Waldo AL, DiMarco JP, for the Atrial FibrillationFollow-up Investigation of Rhythm Management (AFFIRM)Investigators. A comparison of rate control and rhythmcontrol in patients with atrial fibrillation. N Engl J Med2002;347:1825–33.

19. Corley SD, Epstein AE, DiMarco JP, for the AFFIRMInvestigators. Relationships between sinus rhythm, treatment,and survival in the atrial fibrillation follow-up investigation ofrhythm management (AFFIRM) study. Circulation2004;109:1509–13.

20. Van Gelder IC, Hagens VE, Bosker HA, et al. A comparisonof rate control and rhythm control in patients with recurrentpersistent atrial fibrillation. N Engl J Med 2002;347:1834–40.

21. Straus SMJM, Sturkenboom MCJM, Bleumink GS, et al.Noncardiac QTc–prolonging drugs and the risk of suddencardiac death. Eur Heart J 2005;26:2007–12.

696

Page 14: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

22. Ray WA, Murray KT, Meredith S, Narasimhulu SS, Hall K,Stein CM. Oral erythromycin and the risk of sudden deathfrom cardiac causes. N Engl J Med 2004;351:1089–96.

23. Letsas KP, Sideris A, Kounas SP, Efremidis M,Korantzopoulos P, Kardaras F. Drug-induced QT intervalprolongation after ciprofloxacin administration in a patientreceiving olanzapine. Int J Cardiol 2006;109:273–4.

24. Kmierczak J, Peregud-Pogorzelska M, Rzeuski R. QT intervalprolongation and torsades de pointes due to acoadministration of ciprofloxacin and azimilide in a patientwith implantable cardioverter-defibrillator. Pacing ClinElectrophysiol 2007;30:1043–6.

25. Hill SL, Evangelista J-aK, Pizzi AM, Mobassaleh M, FultonDR, Berul CI. Proarrhythmia associated with cisapride inchildren. Pediatrics 1998;101:1053–6.

26. Piquette RK. Torsade de pointes induced by cisapride/clarithromycin interaction. Ann Pharmacother 1999;33:22–6.

27. Roden DM, Balser JR, George AL Jr, Anderson ME. Cardiacion channels. Annu Rev Physiol 2002;64:431–75.

28. Antzelevitch C . Drug-induced spatial dispersion ofrepolarization. Cardiol J 2008;15:100–21.

29. Antzelevitch C, Belardinelli L, Zygmunt AC, et al .Electrophysiological effects of ranolazine, a novel antianginalagent with antiarrhythmic properties. Circulation 2004;110:904–10.

30. Roden DM . Taking the “idio” out of “idiosyncratic”:predicting torsades de pointes. Pacing Clin Electrophysiol1998;21:1029–34.

31. Roden DM. Pharmacogenomics of cardiac arrhyhthmias andimpact on drug therapy. In: Zipes D, Jalife J, eds. Cardiacelectrophysiology: from cell to bedside. Philadelphia:Saunders, 2004:471–8.

32. Yetkin E, Senen K, Ileri M, et al. Diurnal variation of QTdispersion in patients with and without coronary arterydisease. Angiology 2001;52:311–16.

33. Zeltser D, Justo D, Halkin A, Prokhorov V, Heller K, ViskinS. Torsade de pointes due to noncardiac drugs: most patientshave easily identifiable risk factors. Medicine (Baltimore)2003;82:282–90.

34. Yang T, Snyders DJ, Roden DM . Rapid inactivationdetermines the rectification and [K+]o dependence of therapid component of the delayed rectifier K+ current in cardiaccells. Circ Res 1997;80:782–9.

35. Zhang S, Sawanobori T, Adaniya H, Hirano Y, Hiraoka M.Dual effects of external magnesium on action potentialduration in guinea pig ventricular myocytes. Am J PhysiolHeart Circ Physiol 1995;268:H2321–8.

36. McBride BF, Min B, Kluger J, et al. An evaluation of theimpact of oral magnesium lactate on the corrected QT intervalof patients receiving sotalol or dofetilide to prevent atrial orventricular tachyarrhythmia recurrence. Ann NoninvasiveElectrocardiol 2006;11:163–9.

37. Haigney MC, Berger R, Schulman S, et al. Tissue magnesiumlevels and the arrhythmic substrate in humans. J CardiovascElectrophysiol 1997;8:980–6.

38. Rodriguez I, Kilborn MJ, Liu X-K, Pezzullo JC, Woosley RL.Drug-induced QT prolongation in women during themenstrual cycle. JAMA 2001;285:1322–6.

39. Gowda RM, Khan IA, Punukollu G, Vasavada BC, Sacchi TJ,Wilbur SL. Female preponderance in ibutilide-inducedtorsade de pointes. Int J Cardiol 2004;95:219–22.

40. Drici M-D, Knollmann BC, Wang W-X, Woosley RL. Cardiacactions of erythromycin: influence of female sex. JAMA1998;280:1774–6.

41. Makkar RR, Fromm BS, Steinman RT, Meissner MD,Lehmann MH. Female gender as a risk factor for torsades depointes associated with cardiovascular drugs. JAMA1993;270:2590–7.

42. Nerbonne JM, Kass RS. Molecular physiology of cardiacrepolarization. Physiol Rev 2005;85:1205–53.

43. Hondeghem LM, Carlsson L, Duker G. Instability andtriangulation of the action potential predict seriousproarrhythmia, but action potential duration prolongation is

antiarrhythmic. Circulation 2001;103:2004–13.44. Dubin D. Recording the EKG. In: Rapid interpretaion of

EKG’s, 6th ed. Hong Kong: Cover Publications, Inc.,2000:31–54.

45. Extramiana F, Antzelevitch C . Amplified transmuraldispersion of repolarization as the basis for arrhythmogenesisin a canine ventricular-wedge model of short-QT syndrome.Circulation 2004;110:3661–6.

46. Milberg P, Ramtin S, Mönnig G, et al. Comparison of the invitro electrophysiologic and proarrhythmic effects ofamiodarone and sotalol in a rabbit model of acute atrio-ventricular block. J Cardiovasc Pharmacol 2004;44:278–86.

47. van Opstal JM, Schoenmakers M, Verduyn SC, et al. Chronicamiodarone evokes no torsade de pointes arrhythmias despiteQT lengthening in an animal model of acquired long-QTsyndrome. Circulation 2001;104:2722–7.

48. Moro S, Ferreiro M, Celestino D, Medei E, Elizari MV,Sicouri S. In vitro effects of acute amiodarone and dronedaroneon epicardial, endocardial, and M cells of the canine ventricle.J Cardiovasc Pharmacol Ther 2007;12:314–21.

49. Drouin E, Lande G, Charpentier F. Amiodarone reducestransmural heterogeneity of repolarization in the humanheart. J Am Coll Cardiol 1998;32:1063–7.

50. Knollmann BC, Casimiro MC, Katchman AN, et al .Isoproterenol exacerbates a long QT phenotype in Kcnq1-deficient neonatal mice: possible roles for human-like KCNQ1isoform 1 and slow delayed rectifier K+ current. J PharmacolExp Ther 2004;310:311–18.

51. Jost N, Virag L, Bitay M, et al. Restricting excessive cardiacaction potential and QT prolongation: a vital role for IKs inhuman ventricular muscle. Circulation 2005;112:1392–9.

52. Shimizu W, Antzelevitch C. Cellular basis for the ECGfeatures of the LQT1 form of the long–QT syndrome : effectsof �-adrenergic agonists and antagonists and sodium channelblockers on transmural dispersion of repolarization andtorsade de pointes. Circulation 1998;98:2314–22.

53. Zeng J, Rudy Y. Early afterdepolarizations in cardiac myocytes:mechanism and rate dependence. Biophys J 1995;68:949–64.

54. Kozhevnikov DO, Yamamoto K, Robotis D, Restivo M, El-Sherif N. Electrophysiological mechanism of enhancedsusceptibility of hypertrophied heart to acquired torsade depointes arrhythmias: tridimensional mapping of activationand recovery patterns. Circulation 2002;105:1128–34.

55. El-Sherif N, Caref EB, Yin H, Restivo M. The electrophysio-logical mechanism of ventricular arrhythmias in the long QTsyndrome: tridimensional mapping of activation and recoverypatterns. Circ Res 1996;79:474–92.

56. Hryshko L. Membrane pumps and exchangers. In: Zipes D,Jalife J, eds. Cardiac electrophysiology: from cell to bedside,4th ed. Philadelphia: Saunders, 2004:47–8.

57. Watanabe Y, Kimura J. Inhibitory effect of amiodarone onNa+/Ca2+ exchange current in guinea-pig cardiac myocytes. BrJ Pharmacol 2000;131:80–4.

58. Watanabe Y, Kimura J . Acute inhibitory effect ofdronedarone, a noniodinated benzofuran analogue ofamiodarone, on Na+/Ca2+ exchange current in guinea pigcardiac ventricular myocytes. Naunyn Schmiedebergs ArchPharmacol 2008;377:371–6.

59. The ICH Expert Working Group. The clinical evaluation ofQT/QTc interval prolongation and proarrhythmic potential fornon-antiarrhythmic drugs. International conference onharmonisation of technical requirements for registration ofpharmaceuticals for human use, 2005. Available fromhttp://www.ich.org/LOB/media/MEDIA1476.pdf. AccessedOctober 13, 2008.

60. Indik JH, Pearson EC, Fried K, Woosley RL. Bazett andFridericia QT correction formulas interfere with measurementof drug-induced changes in QT interval. Heart Rhythm2006;3:1003–7.

61. Sagie A, Larson M, Goldberg R, Bengtson J, Levy D. Animproved method for adjusting the QT interval for heart rate(the Framingham heart study). Am J Cardiol 1992;70:797–801.

697

Page 15: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

62. Yan G-X, Antzelevitch C. Cellular basis for the normal Twave and the electrocardiographic manifestations of the long-QT syndrome. Circulation 1998;98:1928–36.

63. Viskin MDS, Heller MDK, Barron MDHV, et al .Postextrasystolic U wave augmentation, a new marker ofincreased arrhythmic risk in patients without the long QTsyndrome. J Am Coll Cardiol 1996;28:1746–52.

64. Kirchhof P, Franz MR, Bardai A, Wilde AM. Giant T-U wavesprecede torsades de pointes in long QT syndrome: asystematic electrocardiographic analysis in patients withacquired and congenital QT prolongation. J Am Coll Cardiol2009;54:143–9.

65. Moss A, Schwartz P, Crampton R, et al. The long QTsyndrome: prospective longitudinal study of 328 families.Circulation 1991;84:1136–44.

66. Bednar M, Harrigan EP, Anziano RJ, Camm AJ, Ruskin JN.The QT interval. Prog Cardiovasc Dis 2001;43:1–45.

67. European Heart Rhythm Association, Heart Rhythm Society,Fuster V, et al. ACC/AHA/ESC 2006 guidelines for themanagement of patients with atrial fibrillation—executivesummary: a report of the American College of Cardiology/American Heart Association task force on practice guidelinesand the European Society of Cardiology committee forpractice guidelines (writing committee to revise the 2001guidelines for the management of patients With atrialfibrillation). J Am Coll Card 2006;48:854–906. (Erratum in JAm Coll Cardiol 2007;50:562.)

68. Nademanee K, Schwab MC, Kosar EM, et al. Clinicaloutcomes of catheter substrate ablation for high-risk patientswith atrial fibrillation. J Am Coll Cardiol 2008;51:843–9.

69. Miyasaka Y, Barnes ME, Bailey KR, et al. Mortality trends inpatients diagnosed with first atrial fibrillation: a 21-yearcommunity-based study. J Am Coll Cardiol 2007;49:986–92.

70. Roy D, Talajic M, Nattel S, et al. Rhythm control versus ratecontrol for atrial fibrillation and heart failure. N Engl J Med2008;358:2667–77.

71. Hohnloser SH, Kuck KH, Lilienthal J. Rhythm or rate controlin atrial fibrillation: pharmacological intervention in atrialfibrillation (PIAF)—a randomised trial. Lancet 2000;356:1789–94.

72. Wazni OM, Marrouche NF, Martin DO, et al. Radiofrequencyablation vs antiarrhythmic drugs as first-line treatment ofsymptomatic atrial fibrillation: a randomized trial. JAMA2005;293:2634–40.

73. Stabile G, Bertaglia E, Senatore G, et al. Catheter ablationtreatment in patients with drug-refractory atrial fibrillation: aprospective, multi-centre, randomized, controlled study(catheter ablation for the cure of atrial fibrillation study). EurHeart J 2006;27:216–21.

74. Roux J. Antiarrhythmics after ablation of atrial fibrillation(5A) study. Late-breaking clinical trials session. Presented atthe Heart Rhythm Society 2008 scientific sessions, SanFrancisco, CA, May 15, 2008.

75. Schron EB, Exner DV, Yao Q, et al. Quality of life in theantiarrhythmics versus implantable defibrillators trial: impactof therapy and influence of adverse symptoms anddefibrillator shocks. Circulation 2002;105:589–94.

76. Irvine J, Dorian P, Baker B, et al. Quality of life in theCanadian implantable defibrillator study (CIDS). Am Heart J2002;144:282–9.

77. Kamphuis HCM, de Leeuw JRJ, Derksen R, Hauer RNW,Winnubst JAM. Implantable cardioverter defibrillatorrecipients: quality of life in recipients with and without ICDshock delivery: a prospective study. Europace 2003;5:381–9.

78. Mazur A, Anderson ME, Bonney S, Roden DM. Pause-dependent polymorphic ventricular tachycardia during long-term treatment with dofetilide: a placebo-controlled,implantable cardioverter-defibrillator-based evaluation. J AmColl Cardiol 2001;37:1100–5.

79. Pacifico A, Hohnloser SH, Williams JH, et al. Prevention ofimplantable-defibrillator shocks by treatment with sotalol. NEngl J Med 1999;340:1855–62.

80. Stambler BS, Wood MA, Ellenbogen KA, Perry KT,

Wakefield LK, VanderLugt JT. Efficacy and safety of repeatedintravenous doses of ibutilide for rapid conversion of atrialflutter or fibrillation. Circulation 1996;94:1613–21.

81. Zhang N, Guo JH, Zhang HCh, Li XB, Zhang P, Xn YN.Comparison of intravenous ibutilide vs. propafenone for rapidtermination of recent onset atrial fibrillation. Int J Clin Pract2005;59:1395–400.

82. Fragakis N, Papadopoulos N, Papanastasiou S, et al. Efficacyand safety of ibutilide for cardioversion of atrial flutter andfibrillation in patients receiving amiodarone or propafenone.Pacing Clin Electrophysiol 2005;28:954–61.

83. Glatter K, Yang Y, Chatterjee K, et al. Chemical cardioversionof atrial fibrillation or flutter with ibutilide in patientsreceiving amiodarone therapy. Circulation 2001;103:253–7.

84. Chen YJ, Hsieh MH, Chiou CW, Chen SA . Electro-pharmacologic characteristics of ventricular proarrhythmiainduced by ibutilide. J Cardiovasc Pharmacol 1999;34:237–47.

85. Buchanan LV, Kabell G, Brunden MN, Gibson JK .Comparative assessment of ibutilide, D-sotalol, clofilium, E-4031, and UK-68,798 in a rabbit model of proarrhythmia. JCardiovasc Pharmacol 1993;22:540–9.

86. McBride BF, Yang T, Roden DM. Influence of the G2677T/C3435T haplotype of MDR1 on P-glycoprotein trafficking andibutilide-induced block of HERG. Pharmacogenomics J2009;9:194–201.

87. Singh BN, Singh SN, Reda DJ, et al. Amiodarone versussotalol for atrial fibrillation. N Engl J Med 2005;352:1861–72.

88. Benditt DG, Williams JH, Jin J, et al. Maintenance of sinusrhythm with oral d,l-sotalol therapy in patients withsymptomatic atrial fibrillation and/or atrial flutter. Am JCardiol 1999;84:270–7.

89. Groh WJ, Gibson KJ, McAnulty JH, Maylie JG. �-Adrenergicblocking property of dl-sotalol maintains class III efficacy inguinea pig ventricular muscle after isoproterenol. Circulation1995;91:262–4.

90. Singh S, Zoble RG, Yellen L, et al. Efficacy and safety of oraldofetilide in converting to and maintaining sinus rhythm inpatients with chronic atrial fibrillation or atrial flutter: thesymptomatic atrial fibrillation investigative research ondofetilide (SAFIRE–D) study. Circulation 2000;102:2385–90.

91. Bayer Healthcare Pharmaceuticals. Betapace AF (sotalol)prescribing information. Wayne, NJ; 2007. Available fromhttp://www.betapaceaf.com/PDF/BetapaceAF_PI.pdf. AccessedNovember 17, 2008.

92. Pfizer, Inc. Tikosyn (dofetilide) package insert. New York,NY; 2008. Available from http://www.tikosyn.com/pdf/PI_tikosyn.pdf. Accessed November 17, 2008.

93. Allen MJ, Nichols DJ, Oliver SD. The pharmacokinetics andpharmacodynamics of oral dofetilide after twice daily andthree times daily dosing. Br J Clin Pharmacol 2000;50:247–53.

94. Jonker DM, Kenna LA, Leishman D, Wallis R, Milligan PA,Jonsson EN. A pharmacokinetic-pharmacodynamic model forthe quantitative prediction of dofetilide clinical QTprolongation from human ether-a-go-go–related gene currentinhibition data. Clin Pharmacol Ther 2005;77:572–82.

95. Wu Y, Carlsson L, Liu T, Kowey Pr, Yan G-X. Assessment ofthe proarrhythmic potential of the novel antiarrhythmic agentAZD7009 and dofetilide in experimental models of torsadesde pointes. J Cardiovasc Electrophysiol 2005;16:898–904.

96. Kirchhof P, Milberg P, Eckardt L, Breithardt GN, HaverkampW. Effect of sotalol and acute ventricular dilatation on actionpotential duration and dispersion of repolarization afterdefibrillation shocks. J Cardiovasc Pharmacol 2003;41:640–8.

97. Kaye DM, Dart AM, Jennings GL, Esler MD. Antiadrenergiceffect of chronic amiodarone therapy in human heart failure. JAm Coll Cardiol 1999;33:1553–9.

98. Lalevée N, Barrère-lemaire S, Gautier P, Nargeot J, RichardS. Effects of amiodarone and dronedarone on voltage-dependent sodium current in human cardiomyocytes. JCardiovasc Electrophysiol 2003;14:885–90.

99. Sato R, Koumi S, Singer DH, et al. Amiodarone blocks theinward rectifier potassium channel in isolated guinea pig

698

Page 16: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

ventricular cells. J Pharmacol Exp Ther 1994;269:1213–19.100. Le Bouter S, El Harchi A, Marionneau C, et al. Long-term

amiodarone administration remodels expression of ionchannel transcripts in the mouse heart. Circulation2004;110:3028–35.

101. Kober L, Torp-Pedersen C, McMurray JJV, et al. Increasedmortality after dronedarone therapy for severe heart failure. NEngl J Med 2008;358:2678–87.

102. Singh BN, Connolly SJ, Crijns HJGM, et al. Dronedarone formaintenance of sinus rhythm in atrial fibrillation or flutter. NEngl J Med 2007;357:987–99.

103. Sicouri S, Glass A, Belardinelli L, Antzelevitch C .Antiarrhythmic effects of ranolazine in canine pulmonary veinsleeve preparations. Heart Rhythm 2008;5:1019–26.

104. Kumar K, Nearing B, Bartoli C, Kwaku K, Belardinelli L,Verrier R. Effect of ranolazine on ventricular vulnerabilityand defibrillation threshold in the intact porcine heart. JCardiovasc Electrophysiol 2008;19:1073–9.

105. Chaitman BR, Pepine CJ, Parker JO, et al. Effects ofranolazine with atenolol, amlodipine, or diltiazem on exercisetolerance and angina frequency in patients with severechronic angina: a randomized controlled trial. JAMA2004;291:309–16.

106. Scirica BM, Morrow DA, Hod H, et al. Effect of ranolazine,an antianginal agent with novel electrophysiologicalproperties on the incidence of arrhythmias in patients withnon ST-segment elevation acute coronary syndrome: resultsfrom the metabolic efficiency with ranolazine for lessischemia in non ST-elevation acute coronary syndromethrombolysis in myocardial infarction 36 (MERLIN-TIMI 36)randomized controlled trial. Circulation 2007;116:1647–52.

107. Duff H, Roden D, Primm R, Oates J, Woosley R. Mexiletinein the treatment of resistant ventricular arrhythmias:enhancement of efficacy and reduction of dose-related sideeffects by combination with quinidine. Circulation1983;67:1124–8.

108. Chézalviel-Guilbert F, Davy J-M, Poirier J-M, WeissenburgerJ. Mexiletine antagonizes effects of sotalol on QT intervalduration and its proarrhythmic effects in a canine model oftorsade de pointes. J Am Coll Cardiol 1995;26:787–92.

109. Frederique C-G, Deplanne V, Davy J-M, et al. Combinationof sotalol and quinidine in a canine model of torsades depointes. J Cardiovasc Electrophysiol 1998;9:498–507.

110. Caron MF, Kluger J, Tsikouris JP, Ritvo A, Kalus JS, WhiteCM. Effects of intravenous magnesium sulfate on the QTinterval in patients receiving ibutilide. Pharmacotherapy2003;23: 296–300.

111. Sueta CA, Clarke SW, Dunlap SH, et al. Effect of acutemagnesium administration on the frequency of ventriculararrhythmia in patients with heart failure. Circulation1994;89:660–6.

112. Coleman C, Kalus J, Caron M, Kluger J, White C. Model ofeffect of magnesium prophylaxis on frequency of torsades depointes in ibutilide-treated patients. Am J Health Syst Pharm2004;61:685–8.

113. American Heart Association . 2005 American HeartAssociation guidelines for cardiopulmonary resuscitation andemergency cardiovscular care. Part 7.2. Management ofcardiac arrest. Circulation 2005;112(suppl 1):IV-58–66.

114. Malfatto G, Zaza A, Forster M, Sodowick B, Danilo PJ,Rosen M. Electrophysiologic, inotropic and antiarrhythmiceffects of propafenone, 5-hydroxypropafenone and N-depropylpropafenone. J Pharmacol Exp Ther 1988;246:419–26.

115. Kochiadakis GE, Igoumenidis NE, Hamilos ME, et al. Sotalolversus propafenone for long-term maintenance of normalsinus rhythm in patients with recurrent symptomatic atrialfibrillation. Am J Cardiol 2004;94:1563–6.

116. Bellandi F, Simonetti I, Leoncini M, et al. Long-term efficacyand safety of propafenone and sotalol for the maintenance ofsinus rhythm after conversion of recurrent symptomatic atrialfibrillation. Am J Cardiol 2001;88:640–5.

117. Merot J, Charpentier F, Poirier J-M, Coutris G, Weissenburger

J. Effects of chronic treatment by amiodarone on transmuralheterogeneity of canine ventricular repolarization in vivo:interactions with acute sotalol. Cardiovasc Res1999;44:303–14.

118. Darke S, Kaye S, Duflou J. Comparative cardiac pathologyamong deaths due to cocaine toxicity, opioid toxicity andnon-drug-related causes. Addiction 2006;101:1771–7.

119. Puigdollers E, Domingo-Salvany A, Brugal T, et al .Characteristics of heroin addicts entering methadonemaintenance treatment: quality of life and gender. Subst UseMisuse 2004;39:1353–68.

120. Berkman L, Blummenthal J, Burg M, et al. Effects of treatingdepression and low perceived social support on clinical eventsafter myocardial infarction: the enhancing recovery incoronary heart disease patients (ENRICHD) randomized trial.JAMA 2003;289:3106–16.

121. Ferketich AK, Schwartzbaum JA, Frid DJ, MoeschbergerML. Depression as an antecedent to heart disease amongwomen and men in the NHANES I study. Arch Intern Med2000;160:1261–8.

122. van Melle JP, de Jonge P, Honig A, et al . Effects ofantidepressant treatment following myocardial infarction.British J Psychiatry 2007;190:460–6.

123. Bär K-J, Koschke M, Boettger MK, et al. Acute psychosisleads to increased QT variability in patients suffering fromschizophrenia. Schizophr Res 2007;95:115–23.

124. Bobes J, Arango C, Aranda P, Carmena R, Garcia-Garcia M,Rejas J. Cardiovascular and metabolic risk in outpatients withschizophrenia treated with antipsychotics: results of theCLAMORS study. Schizophr Res 2007;90:162–73.

125. Cohn T, Prud’homme D, Steiner D, Kameh H, Remington G.Characterizing coronary heart disease risk in chronicschizophrenia: high prevalence of the metabolic syndrome.Can J Psychiatry 2004;49:753–60.

126. Saari K, Linderman S, Viilo K, et al. A 4-fold risk ofmetabolic syndrome in patients with schizophrenia: theNorthern Finland 1966 birth cohort study. J Clin Psychiatry2005;66:559–63.

127. Stajer D, Mozina H, Noc M, Rode P. Correlation betweenQTc interval duration and left ventricular systolic dysfunctionin patients with acute myocardial infarction. J Electrocardiol1993;26:333–40.

128. Eap CB, Crettol S, Rougier JS, et al. Stereoselective block ofhERG channel by (S)-methadone and QT intervalprolongation in CYP2B6 slow metabolizers. Clin PharmacolTher 2007;81:719–28.

129. Krantz MJ, Lowery CM, Martell BA, Gourevitch MN,Arnsten JH. Effects of methadone on QT-interval dispersion.Pharmacotherapy 2005;25:1523–9.

130. Krantz MJ, Martin J, Stimmel B, Mehta D, Haigney MCP.QTc interval screening in methadone treatment. Ann InternMed 2009;150:387–95.

131. Wedam EF, Bigelow GE, Johnson RE, Nuzzo PA, HaigneyMCP. QT-interval effects of methadone, levomethadyl, andbuprenorphine in a randomized trial. Arch Intern Med2007;167:2469–75.

132. Oviedo-Joekes E, Brissette S, Marsh DC, et al .Diacetylmorphine versus methadone for the treatment ofopioid addiction. N Engl J Med 2009;361:777–86.

133. Moore TA, Buchanan RW, Buckley PF, et al. The Texasmedication algorithm project antipsychotic algorithm forschizophrenia: 2006 update. J Clin Psychiatry 2007;68:1751–62.

134. Marder S. A review of agitation in mental illness: treatmentguidelines and current therapies. J Clin Psychiatry 2006;67(suppl 10):13–21.

135. Crumb W, Ekins S, Sarazan R, et al. Effects of antipsychoticdrugs on Ito ,INa,Isus,IK1, and hERG: QT prolongation,structure activity relationship , and network analysis. PharmRes 2006;23:1133–43.

136. Kongsamut S, Kang J, Chen X-L, Roehr J, Rampe D. Acomparison of the receptor binding and HERG channelaffinities for a series of antipsychotic drugs. Eur J Pharmacol2002;450:37–41.

699

Page 17: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

PHARMACOTHERAPY Volume 30, Number 7, 2010

137. Anonymous. Study 054. Briefing document for Zeldoxcapsules (ziprasidone HCL). FDA psychopharmacologicaldrugs advisory committee, July 19, 2000. Available fromhttp:// www.fda.gov/ohrms/dockets/ac/00/backgrd/3619b1a.pdf.

138. Thomas D, Wu K, Kathofer S, et al. The antipsychotic drugchlorpromazine inhibits HERG potassium channels. Br JPharmacol 2003;139:567–74.

139. Brown CS, Farmer RG, Soberman JE, Eichner SF .Pharmacokinetic factors in the adverse cardiovascular effectsof antipsychotic drugs. Clin Pharmacokinet 2004;43:33–56.

140. Mörl D, Agneter E, Krivanek P, Koppatz K, Todt H. Dual ratedependent cardiac electrophysiological effects of haloperidol:slowing of intraventricular conduction and lengthening ofrepolarization. J Cardiovasc Pharmacol 2003;41:870–9.

141. Hoehns JD, Stanford RH, Geraets DR, Skelly KS, Lee HC,Gaul BL. Torsades de pointes associated with chlorpromazine:case report and review of associated ventricular arrhythmias.Pharmacotherapy 2001;21:871–83.

142. Tisdale JE, Rasty S, Padhi ID, Sharma ND, Rosman H. Theeffect of intravenous haloperidol on QT interval dispersion incritically ill patients: comparison with QT intervalprolongation for assessment of risk of torsades de pointes. JClin Pharmacol 2001;41:1310–18.

143. Dorado P, Berecz R, Penas-Lledo EM, Llerena A .Antipsychotic drugs and QTc prolongation: the potential roleof CYP2D6 genetic polymorphism. Expert Opin Drug MetabToxicol 2007;3:9–19.

144. O’Brien JM, Rockwood RP, Suh KI. Haloperidol-inducedtorsade de pointes. Ann Pharmacother 1999;33:1046–50.

145. Perrault LP, Denault AY, Carrier M, Cartier R, Belisle S.Torsades de pointes secondary to intravenous haloperidolafter coronary bypass grafting surgery. Can J Anesth2000;47:251–4.

146. Sharma ND, Rosman HS, Padhi ID, Tisdale JE. Torsades depointes associated with intravenous haloperidol in critically illpatients. Am J Cardiol 1998;81:238–40.

147. Czekalla J, Beasley C, Dellva M, Berg P, Grundy S. Analysisof the QTc interval during olanzapine treatment of patientswith schizophrenia and related psychosis. J Clin Psychiatry2001;62:191–8.

148. Milnes JT, Witchel HJ, Leaney JL, Leishman DJ, Hancox JC.hERG K+ channel blockade by the antipsychotic drugthioridazine: an obligatory role for the S6 helix residue F656.Biochem Biophys Res Commun 2006;351:273–80.

149. Marder SR, McQuade RD, Stock E, et al. Aripiprazole in thetreatment of schizophrenia: safety and tolerability in short-term, placebo-controlled trials. Schizophr Res 2003;61:123–36.

150. Furst BA, Champion KM, Pierre JM, Wirshing DA, WirshingWC. Possible association of QTc interval prolongation withco-administration of quetiapine and lovastatin. BiologicPsychiatry 2002;51:264–5.

151. Vieweg WVR, Schneider RK, Ma W. Torsade de pointes in apatient with complex medical and psychiatric conditionsreceiving low-dose quetiapine. Acta Psychiatr Scand2005;112:318–22.

152. Raviña T, Raviña P, Gutierrez J. Acquired long QT syndrome:risperidone-facilitated triggered activity and torsades depointes during complete AV block. Int J Cardiol 2007;116:416–20.

153. Llerena A, Berecz R, Dorado P, de la Rubia A. QTc interval,CYP2D6 and CYP2C9 genotypes and risperidone plasmaconcentrations. J Psychopharmacol 2004;18:189–93.

154. Vieweg W. New generation antipsychotic drugs and QTcinterval prolongation. Prim Care Companion J ClinPsychiatry 2003;5:205–15.

155. Simko J, Csilek A, Karaszi J, Lorincz I. Proarrhythmicpotential of antimicrobial agents. Infection 2008;36:194–206.

156. Shaffer D, Singer S, Korvick J, Honig P. Concomitant riskfactors in reports of torsades de pointes associated withmacrolide use: review of the United States Food and DrugAdministration adverse event reporting system. Clin InfectDis 2002;35:197–200.

157. Justo D, Zeltser D . Torsades de pointes induced by

antibiotics. Eur J Intern Med 2006;17:254–9.158. Nattel S, Ranger S, Talajic M, Lemery R, Roy D .

Erythromycin-induced long QT syndrome: concordance withquinidine and underlying cellular electrophysiologicmechanism. Am J Med 1990;89:235–8.

159. Stanat SJ, Carlton CG, Crumb WJ Jr, Agrawal KC, ClarksonCW . Characterization of the inhibitory effects oferythromycin and clarithromycin on the HERG potassiumchannel. Mol Cell Biochem 2003;254:1–7.

160. Volberg WA, Koci BJ, Su W, Lin J, Zhou J. Blockade ofhuman cardiac potassium channel human ether-a-go-go-related gene (HERG) by macrolide antibiotics. J PharmacolExp Ther 2002;302:320–7.

161. Rubart M, Pressler M, Pride H, Zipes D. Electrophysiologicalmechanisms in a canine model of erythromycin-associatedlong QT syndrome. Circulation 1993;88:1832–44.

162. Antzelevitch CS, Zhang ZQ, Yan GX. Cellular and ionicmechanisms underlying erythromycin-induced long QTintervals and torsade de pointes. J Am Coll Cardiol1996;28:1836–48.

163. Milberg P, Eckardt L, Bruns HJ, et al . Divergentproarrhythmic potential of macrolide antibiotics despitesimilar QT prolongation: fast phase 3 repolarization preventsearly afterdepolarizations and torsade de pointes. J PharmacolExp Ther 2002;303:218–25.

164. Austin KL, Mather LE, Philpot CR, McDonald PJ .Intersubject and dose-related variability after intravenousadministration of erythromycin. Br J Clin Pharmacol1980;10:273–9.

165. Schoenenberger RA, Haefeli WE, Weiss P, Ritz RF .Association of intravenous erythromycin and potentially fatalventricular tachycardia with Q-T prolongation (torsades depointes). BMJ 1990;300:1375–6.

166. Tschida SJ, Guay DRP, Straka RJ, Hoey LL, Johanning R,Vance-Bryan K. QTc-interval prolongation associated withslow intravenous erythromycin lactobionate infusions incritically ill patients: a prospective evaluation and review ofthe literature. Pharmacotherapy 1996;16:663–74.

167. Lee KL, Jim MH, Tang SC, Tai YT. QT prolongation andtorsades de pointes associated with clarithromycin. Am J Med1998;104:395–6.

168. Kamochi H, Nii T, Eguchi K, et al. Clarithromycin associatedwith torsades de pointes. Jpn Circ J 1999;63:421–2.

169. Desta Z, Kerbusch T, Flockhart DA. Effect of clarithromycinon the pharmacokinetics and pharmacodynamics of pimozidein healthy poor and extensive metabolizers of cytochromeP450 2D6 (CYP2D6). Clin Pharmacol Ther 1999;65:10–20.

170. Flockhart DA, Drici MD, Kerbusch T, et al. Studies on themechanism of a fatal clarithromycin-pimozide interaction in apatient with Tourette syndrome. J Clin Psychopharmacol2000;20:317–24.

171. Hayashi Y, Ikeda U, Hashimoto T, Watanabe T, MitsuhashiT, Shimada K. Torsades de pointes ventricular tachycardiainduced by clarithromycin and disopyramide in the presenceof hypokalemia. Pacing Clin Electrophysiol 1999;22:672–4.

172. Paar D, Terjung B, Sauerbruch T. Life-threatening interactionbetween clarithromycin and disopyramide. Lancet1997;349:326–7.

173. Sekkarie MA. Torsades de pointes in two chronic renal failurepatients treated with cisapride and clarithromycin. Am JKidney Dis 1997;30:437–9.

174. Owens RC Jr. QT prolongation with antimicrobial agents:understanding the significance. Drugs 2004;64:1091–124.

175. Iannini PB, Doddamani S, Byazrova E, Curciumaru I,Kramer H. Risk of torsades de pointes with non-cardiacdrugs: prolongation of QT interval is probably a class effect offluoroquinolones. BMJ 2001;322:46–7.

176. Hagiwara T, Satoh S, Kasai Y, Takasuna K. A comparativestudy of the fluoroquinolone antibacterial agents on theaction potential duration in guinea pig ventricular myocardia.Jpn J Pharmacol 2001;87:231–4.

177. Demolis JL, Kubitza D, Tenneze L, Funck-Brentano C. Effectof a single oral dose of moxifloxacin (400 mg and 800 mg) on

700

Page 18: Drug-Induced QT-Interval Prolongation: Considerations for Clinicians

DRUG-INDUCED QT-INTERVAL PROLONGATION Li et al

ventricular repolarization in healthy subjects. Clin PharmacolTher 2000;68:658–66.

178. Tsikouris JP, Peeters MJ, Cox CD, Meyerrose GE, Seifert CF.Effects of three fluoroquinolones on QT analysis afterstandard treatment courses. Ann Noninvasive Electrocardiol2006;11:52–6.

179. Wassmann S, Nickenig G, Bohm M. Long QT syndrome andtorsade de pointes in a patient receiving fluconazole [casereport]. Ann Intern Med 1999;131:797.

180. Tholakanahalli VN, Potti A, Hanley JF, Merliss AD.Fluconazole-induced torsade de pointes. Ann Pharmacother2001;35:432–4.

181. Khazan M, Mathis AS. Probable case of torsades de pointesinduced by fluconazole. Pharmacotherapy 2002;22:1632–7.

182. Tatetsu H, Asou N, Nakamura M, et al. Torsades de pointesupon fluconazole administration in a patient with acutemyeloblastic leukemia. Am J Hematol 2006;81:366–9.

183. Alkan Y, Haefeli WE, Burhenne J, Stein J, Yaniv I, Shalit I.Voriconazole-induced QT interval prolongation andventricular tachycardia: a non-concentration-dependentadverse effect. Clin Infect Dis 2004;39:e49–52.

184. Philips JA, Marty FM, Stone RM, Koplan BA, Katz JT, BadenLR. Torsades de pointes associated with voriconazole use.Transpl Infect Dis 2007;9:33–6.

185. Eiden C, Peyriere H, Tichit R, et al. Inherited long QTsyndrome revealed by antifungals drug-drug interaction. JClin Pharm Ther 2007;32:321–4.

186. Strevel E, Ing DJ, Siu LL. Molecularly targeted oncologytherapeutics and prolongation of the QT interval. J ClinOncol 2007;25:3362–71.

187. National Cancer Institute. Cancer therapy evaluationprogram, common terminology criteria for adverse events,version 4.0. Available from http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.02_2009–09-15_QuickReference_8.5x11.pdf.

188. Bristol-Myers Squibb. Sprycel (dasatinib) prescribinginformation. Princeton, NJ; 2009.

189. Novartis Pharmaceuticals Corporation. Tasigna (nilotinib)prescribing information. East Hanover, NJ; 2007.

190. Pfizer Inc. Sutent (sunitinib) prescribing information. NewYork, NY; 2008.

191. Buckstein R, Meyer RM, Seymour L, et al. Phase II testing of

sunitinib: the National Cancer Institute of Canada clinicaltrials group IND program trials IND182–185. Curr Oncol2007;14:154–61.

192. National Comprehensive Cancer Network. The NCCNclinical practice guidelines in oncology, acute myeloidleukemia (version 1.2010). Fort Washington, PA: NationalComprehensive Cancer Network, 2009.

193. Soignet SL, Frankel SR, Douer D, et al. United Statesmulticenter study of arsenic trioxide in relapsed acutepromyelocytic leukemia. J Clin Oncol 2001;19:3852–60.

194. Unnikrishnan D, Dutcher JP, Varshneya N, et al. Torsades depointes in 3 patients with leukemia treated with arsenictrioxide. Blood 2001;97:1514–16.

195. Floyd JD, Nguyen DT, Lobins RL, Bashir Q, Doll DC, PerryMC . Cardiotoxicity of cancer therapy. J Clin Oncol2005;23:7685–96.

196. He J, Kargacin ME, Kargacin GJ, Ward CA. Tamoxifeninhibits Na+ and K+ currents in rat ventricular myocytes. AmJ Physiol Heart Circ Physiol 2003;285:H661–8.

197. Liu X-K, Katchman A, Ebert SN, Woosley RL . Theantiestrogen tamoxifen blocks the delayed rectifier potassiumcurrent, IKr, in rabbit ventricular myocytes. J Pharmacol ExpTher 1998;287:877–83.

198. Slovacek L, Ansorgova V, Macingova Z, Haman L, Petera J.Tamoxifen-induced QT interval prolongation. J Clin PharmTher 2008;33:453–5.

199. Jackson CW, Sheehan AH, Reddan JG. Evidence-basedreview of the black-box warning for droperidol. Am J HealthSyst Pharm 2007;64:1174–86.

200. Nuttall GA, Eckerman KM, Jacob KA, et al. Does low-dosedroperidol administration increase the risk of drug-inducedQT prolongation and torsade de pointes in the generalsurgical population? Anesthesiology 2007;107:531–6.

201. Habib AS, Gan TJ. Food and Drug Administration black boxwarning on the perioperative use of droperidol: a review ofthe cases. Anesth Analg 2003;96:1377–9.

202. Merck & Co ., Inc . Zolinza (vorinostat) prescribinginformation. Whitehouse Station, NJ; 2009.

203. Dennis A, Wang L, Wan X, Ficker E . hERG channeltrafficking: novel targets in drug-induced long QT syndrome.Biochem Soc Trans 2007;035:1060–3.

701