Article

Mechanisms Underlying the Actions of Antidepressant and Antipsychotic Drugs That Cause Sudden Cardiac Arrest

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

A number of antipsychotic and antidepressant drugs are known to increase the risk of ventricular arrhythmias and sudden cardiac death. Based largely on a concern over the development of life-threatening arrhythmias, a number of antipsychotic drugs have been temporarily or permanently withdrawn from the market or their use restricted. While many antidepressants and antipsychotics have been linked to QT prolongation and the development of torsade de pointes arrhythmias, some have been associated with a Brugada syndrome phenotype and the development of polymorphic ventricular arrhythmias. This article examines the arrhythmic liability of antipsychotic and antidepressant drugs capable of inducing long QT and/or Brugada syndrome phenotypes. The goal of this article is to provide an update on the ionic and cellular mechanisms thought to be involved in, and the genetic and environmental factors that predispose to, the development of cardiac arrhythmias and sudden cardiac death among patients taking antidepressant and antipsychotic drugs that are in clinical use.

Disclosure:The authors acknowledge financial support from the National Heart, Lung, and Blood Institute (NHLBI) (Grant #HL47678) and from the Martha and Wistar Morris Fund.

Received:

Accepted:

Acknowledgements:The authors acknowledge Gan-Xin Yan, Lin Wu and Subramanian Krishnan’s assistance in providing us with ECGs of patients administered with antipsychotic and antidepressant drugs.

Correspondence Details:Charles Antzelevitch, Professor and Executive Director, Cardiovascular Research, Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA, Director of Research, Lankenau Heart Institute, Wynnewood, PA 19096, USA, E: cantzelevitch@gmail.com

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

The link between sudden unexplained death in individuals with mental health problems who are administered antipsychotic drugs has been recognised for over a century.1 A clear relationship has emerged over the past 25 years between antipsychotic drugs, prolongation of the QT interval of the ECG, atypical polymorphic tachycardia known as torsade de pointes (TdP) and sudden cardiac death (SCD). A number of antipsychotic drugs have been temporarily or permanently withdrawn from the market – or their use restricted – because of a concern over QT and QT corrected for heart rate (QTc) prolongation and development of TdP. In some cases, close follow-up with an ECG has been recommended or a modification of label imposed. The list of antipsychotic drugs implicated includes pimozide, sertindole, thioridazine, mesoridazine, promazine, triflupromazine, droperidol, moperone, pipamperone, sultopride and ziprasidone. A link between clinical use of antidepressants and the development of arrhythmias was first suggested following the Cardiac Arrhythmia Suppression Trial (CAST),2 based on the sodium channel-blocking properties of a number of antidepressant drugs including imipramine. Tricyclic antidepressants were subsequently shown to inhibit potassium channels and thus to prolong the QT interval and induce TdP. These effects of tricyclic antidepressants are generally observed when combined with other QT-prolonging agents or in cases of overdose. In addition, a number of case reports have linked tricyclic antidepressants as well as antipsychotic drugs to drug-induced Brugada syndrome (BrS), leading to syncope and SCD as a result of the development of rapid polymorphic ventricular tachycardia (VT) and VF.

Our focus in this article is on mechanisms and predisposing factors underlying the development of cardiac arrhythmias and SCD associated with antidepressant and antipsychotic drugs in clinical use.

Drug-induced Long QT Syndrome and Torsade de Pointes by Antidepressant and Antipsychotic Agents

The QT interval is a measure of the time interval between the start of depolarisation and the end of repolarisation. QTc intervals above 450 ms in men and 460 ms in women are considered to be abnormally prolonged. TdP, from the French for ‘twisting of the points’, is an atypical VT characterised by oscillations of the points or R wave peaks (‘pointes’) around the main axis of the ECG, giving rise to a unique morphology. Since the original work of François Dessertenne,3 it has been well recognised that many conditions are capable of causing prolonged or abnormal repolarisation, giving rise to QT prolongation, abnormal T/U wave morphologies and the development of TdP.4 Although a prolonged QT interval is essential for the development of TdP, it is generally not considered sufficient to induce TdP. An increased risk for TdP is recognised when the QTc exceeds 500 ms and whenever a drug increases QTc by >60–70 ms, especially when the increase develops rapidly.4–6 In addition to QTc prolongation, the risk of TdP is associated with the dispersion of transmural repolarisation and excitability. TdP arrhythmias can degenerate into VF, leading to SCD.

TdP can be caused by either congenital or acquired long QT syndrome (LQTS). Congenital LQTS is subdivided into 10 genotypes distinguished by mutations in at least 17 different genes encoding for cardiac ion channels and structural anchoring proteins. In the different genotypes, cardiac events may be precipitated by physical or emotional stress (LQT1), a startle (LQT2), or may occur at rest or during sleep (LQT3).

Prolong the QT Interval & Induce Torsade de Pointes

Article image

Acquired LQTS refers to a syndrome caused by circumstances other than genetic factors, including exposure to drugs that prolong the duration of the ventricular action potential (AP),7 or secondary to cardiomyopathies such as dilated or hypertrophic cardiomyopathy, as well as to abnormal QT prolongation associated with bradycardia or electrolyte imbalance.8–13 The acquired form of the disease is far more prevalent than the congenital form, and in some cases may have a genetic predisposition.

Table 1, modified from www.CredibleMeds.org, lists the antidepressant and antipsychotic drugs that have been shown to prolong QT interval and induce TdP. Among the antidepressant agents, amitriptyline, imipramine, and maprotiline are the agents most commonly associated with TdP the greatest prolongation of QT interval is observed with maprotiline).14 Table 1 shows the relative risk of TdP for distinct antidepressants and antipsychotics by classifying the drugs in TdP risk categories ranging from the highest risk of TdP; known risk of TdP (KR), to possible risk of TdP (PR) and conditional risk of TdP (CR). As shown in Table 1, most antidepressants are classified as CR, with a low risk of TdP. Vieweg and Wood14 reported 13 cases of TdP induced by antidepressants. As is typical of acquired LQTS, most cases (12 of 13) involved women. One case involved a child. In addition to female sex, risk factors include age (peaking in adolescence), bradycardia, metabolic inhibitors, hypokalemia, hypomagnesaemia, drug overdose and co-administration of QT-prolonging drugs (Table 2). QRS duration of the ECG, measured in five of 13 cases, showed prolongation in two and no change in three, suggesting that QT prolongation with antidepressants may be a result of sodium as well as potassium channel blockade. In a recent study, Danielsson and colleagues confirmed that antidepressants with KR or PR of TdP were associated with a higher risk than those classified as CR of TdP.15 Selective serotonin reuptake inhibitors (SSRIs), including citalopram, escitalopram, fluoxetine, paroxetine, sertraline and venlafaxine have been shown to exhibit a higher risk than tricyclic antidepressants, especially considering that an increase in dose of SSRIs, but not that of tricyclic antidepressants, markedly increases the risk of sudden death.16 For the most commonly used antidepressants in the elderly, the following risk ranking was observed, from highest to lowest; mirtazapine;citalopram;sertraline;amitriptyline.15 In 2011, the Food and Drug Administration (FDA) issued a warning concerning the antidepressant citalopram (Celexa®, Allergan) and its potential risk for TdP at doses greater than 40 mg (FDA, drug safety communication). Providers were asked to use doses less than 20 mg in patients over 60 years of age with hepatic dysfunction, as well as in poor CYP2C19 metabolisers or those taking concomitant CYP2C19 inhibitors. The maximum daily dosage of citalopram is now 40 mg in the non-elderly adult population. Most studies show that – in contrast to antipsychotics – antidepressants, including SSRIs, induce QT prolongation and TdP arrhythmias in cases of overdose, but rarely at therapeutic concentrations. However, QT prolongation may occur in elderly patients at therapeutic doses.16

Among the newer non-SSRIs, QT prolongation has rarely been reported with venlafaxine at therapeutic doses or with overdose. Bupropion has been linked to QT prolongation in overdose situations. In elderly patients with a number of high-risk comorbidities, mirtazapine demonstrated higher risk of SCD and ventricular arrhythmias than paroxetine.16 Jasiak et al. concluded that, based on the current literature, risk of QT/QTc prolongation with most newer non-SSRI antidepressants at therapeutic doses is low.16 The highest risk for QT prolongation appears to exist in overdose situations with venlafaxine and bupropion. They note that, given the few controlled studies and confounding variables in case reports, it is difficult to draw conclusions on QT prolongation risk with many of the newer non-SSRI antidepressants.

Antipsychotic drugs, especially those in the phenothiazine group, can also induce QT prolongation and TdP. Mehtonen et al. surveyed cases of sudden death by antidepressant or antipsychotic drugs and found 49 cases of sudden death (31 women and 18 men) associated with the use of these agents.17 A therapeutic dose of phenothiazine was involved in 46 of the 49 cases. Thioridazine was the only antipsychotic drug administered in 15 of the 49 cases. Figure 1 displays a case of a marked QT prolongation and TdP induced by the antipsychotic agent flupentixol.

Antipsychotic drugs generally have a higher torsadogenic potential (category KR and PR) than antidepressants (category CR) (Tables 1 and 3). As a consequence, antidepressant-induced TdP is more typically observed in the presence of drug combinations. Ray et al. conducted a retrospective cohort study of half a million Medicaid patients between 1988 and 1993, before the introduction of atypical antipsychotics, and observed that the risk for sudden death increased 2.39 times in individuals receiving antipsychotic drugs compared with those who did not receive these agents.18 Although the study did not demonstrate causality, it suggested that the potential adverse cardiac effects of antipsychotics should be considered in clinical practice, particularly for patients with cardiovascular disease. Hennessy et al. in a study of 90,000 patients, observed that those treated for schizophrenia had a higher incidence of cardiac arrest and ventricular arrhythmias than non-schizophrenia patients.19 The drugs used were clozapine, haloperidol, risperidone, and thioridazine. In a study of nursing home residents in six states, Liperoti et al. observed that the use of conventional antipsychotics led to a twofold increase in risk of hospitalisation for ventricular arrhythmias and cardiac arrest, especially in patients with pre-existing cardiac disease.20 In the elderly, haloperidol has been associated with high risk of TdP.15 The IV form of haloperidol is thought to carry a higher risk of QTc prolongation and TdP than the oral form.21,22 In 11 cases of fatal TdP, eight occurred with IV haloperidol. The FDA now recommends cardiac monitoring for all patients receiving haloperidol.

Drug-induced Brugada Syndrome Phenotype by Antidepressant and Antipsychotic Agents

BrS was introduced as a new clinical entity by Pedro and Josep Brugada in 1992.23 The syndrome has been associated with a high risk of sudden death, especially in men as they enter their third and fourth decades of life. A consensus report published in 2002 delineated diagnostic criteria for the syndrome.24,25 A second consensus conference report published in 2005 focused on risk stratification schemes and approaches to therapy.26,27 The most recent expert consensus report focused on emerging concepts, advances in risk stratification and approaches to therapy for both Brugada and early repolarisation syndrome (ERS), the so-called J wave syndromes.28

BrS is characterised by an electrocardiographic pattern of right bundle brunch in right precordial leads, ST-segment elevation in the right precordial leads, relatively normal QTc interval, coupled with syncope and sudden death caused by VT/VF in patients with no or minimal structural disease.

Rick Factors for Torsade de Pointes and Brugada Syndrome

Article image

Flupentixol

Article image

Three types of repolarisation patterns in the right precordial leads are recognised.24,25 Type 1 ST-segment elevation is diagnostic of BrS and is characterised by a coved ST-segment elevation ≥2 mm (0.2 mV) followed by a negative T wave. Type 2 ST-segment elevation has a saddleback appearance with a high take-off ST-segment elevation of ≥2 mm, followed by a trough displaying ≥1 mm ST elevation, followed by either a positive or biphasic T wave. Type 3 ST-segment elevation has either a saddleback or coved appearance with an ST-segment elevation of <1 mm. These three patterns may be observed sequentially in the same patient or following the introduction of specific drugs, particularly sodium channel blockers. Type 2 and type 3 ST-segment elevation are not considered to be diagnostic of BrS. BrS is definitively diagnosed only when a type 1 ST-segment elevation (Brugada ECG) is observed in more than one right-precordial lead (V1–V3), in the presence or absence of sodium channel-blocking agent, and in conjunction with one or more of the following; documented VF, polymorphic VT; a family history of SCD (<45 years old); coved type ECGs in family members; inducibility of VT with programmed electrical stimulation; syncope; or nocturnal agonal respiration.24–27

The average age at the time of cardiac arrest in patients with BrS is approximately 45 years, but most develop symptoms between 20 and 65 years.29–31 BrS in children is rare, but sudden death in this population is reported.32,33 Men are at increased risk for development of a spontaneous type I Brugada ECG and SCD.34,35

Durgs that Prolong the QT Interval and induce Torsade de Pointes

Article image

In a recent study, women comprised 42 % of a cohort of 542 patients who presented with a spontaneous or drug-induced Brugada type 1 pattern on the ECG.36 Women with BrS present more benign clinical characteristics, less spontaneous type 1 ECG pattern, and are more likely to be asymptomatic than men.35

BrS has been associated with variants in 19 different genes. The gene most often associated with BrS is SCN5A, accounting for 11–28 % of cases, depending largely on geographic location. Over 300 BrS-related variants in SCN5A, the gene encoding the alpha subunit of the cardiac sodium channel, have been reported.37–40 Loss-of-function mutations in SCN5A contribute to the development of both BrS and ERS, as well as to various conduction diseases, Lenegre’s disease and sick sinus syndrome.

Variants in genes encoding the calcium channels including CACNA1C (Cav1.2), CACNB2b (Cavbeta2b) and CACNA2D1 (Cavalpha2delta) have been reported in up to 13 % of probands.41–44 Mutations in glycerol-3-phosphate dehydrogenase 1-like enzyme gene (GPD1L), SCN1B (beta1-subunit of sodium channel), KCNE3 (MiRP2), SCN3B (beta3-subunit of soduim channel), KCNJ8 (Kir6.1), KCND3 (Kv4.3), RANGRF (MOG1), SLMAP, ABCC9 (SUR2A), (Navbeta2), PKP2 (plakophillin-2), FGF12 (FHAF1), HEY2, SEMA3A (semaphorin) and KCNAB2 (Kvbeta2) are relatively rare.45–56 An association of BrS with SCN10A, a gene encoding a neuronal sodium channel, was first reported in 2014.56–58 There is controversy as to the pathogenicity of many SCN10A mutations with yields ranging from 5.0 % to 16.7 %.57–59 Mutations in all of these genes lead to loss of function in sodium (INa) and calcium (ICa) channel currents, as well as to a gain of function in transient outward potassium current (Ito) or ATP-sensitive potassium current (IK-ATP).58,60

New susceptibility genes proposed and awaiting confirmation include the transient receptor potential melastatin protein 4 gene (TRPM4)61 and the KCND2 gene. Variants in KCNH2, KCNE5, SEMA3A, although not causative, have been identified as capable of modulating the substrate for the development of BrS.62–65 KCNE4 has recently been added to this group (unpublished observation, Clatot and Antzelevitch). Loss-of-function mutations in HCN4 (prominently expressed in the sinus node) have been associated with BrS but may be modulatory by acting to unmask BrS by reducing heart rate.66

A large number of factors modulate the electrocardiographic and arrhythmic manifestations of BrS. ST-segment elevation in BrS is often dynamic. The Brugada ECG may be concealed, but can be unmasked or modulated by sodium channel blockers, a febrile state, vagotonic agents, alpha adrenergic agonists, beta adrenergic blockers, tricyclic or tetracyclic antidepressants, first generation antihistamines (dimenhydrinate), a combination of glucose and insulin, hyperkalemia, hypokalemia, hypercalcaemia, and by alcohol and cocaine toxicity.67–77 These agents may also induce acquired forms of BrS. Propafenone, typically prescribed for the treatment of AF, is a common example of a drug that can unmask BrS.78–81 Lithium, a widely used antidepressant agent, has been recently added to the list of drugs to avoid in patients with BrS. Lithium is a potent blocker of cardiac sodium channels and can unmask a type 1 ECG in patients with BrS.82 Propofol, a short-acting, IV-administered sedative-hypnotic and antiepileptic agent with anaesthetic properties, may cause a rare condition called propofol infusion syndrome, characterised by unexplained lactic acidosis, lipaemia, rhabdomyolysis, cardiovascular collapse and Brugada-like ECG pattern following high-dose propofol infusion over prolonged periods of time.83,84

Table 3 lists the antidepressants and antipsychotic agents reported to induce the Brugada ECG pattern. All cases of BrS pattern induced by antidepressants and antipsychotics displayed a type 1 ST-segment elevation (Figure 2). A study of 98 patients experiencing an overdose of tricyclic antidepressants reported that 15 of these displayed an ECG consistent with BrS.71 The overall mortality was 3.0 % among all patients, but 6.7 % among patients who displayed a Brugada phenotype. Rouleau et al. described three cases of psychotropic drug-induced Brugada ECG,85 occurring during concomitant administration of amitriptyline and a phenothiazine (case 1), overdose of fluoxetine (case 2), and co-administration of trifluoperazine and loxapine (case 3). Babaliaros and Hurst described a Brugada pattern in patients receiving increasing doses of imipramine.70 Akhtar and Goldschlager reported a case of BrS following massive ingestion of desipramine and clonazepam.77 Chow et al. reported a similar case involving desipramine.86 Bolognesi et al. described a Brugada ECG pattern following overdose of amitriptyline and with maprotiline.87 Additional cases of BrS were reported following overdose with nortriptyline72,88 or lithium.82,89

The available data suggest that most cases of antidepressant and antipsychotic-induced BrS phenotype occur as a consequence of drug overdose or drug combination.

Effects of antidepressants and antipsychotics on ion channels

Antidepressants and antipsychotics are reported to modulate the cardiac AP by blocking a variety of cardiac ion channels. In the ventricle they inhibit the fast sodium channel inward current (INa), the inward calcium current (ICa), and one or more outward potassium currents (IK), particularly the rapidly activating delayed rectifier current (IKr). Drug-induced IKr block has attracted considerable attention in recent years because of the association of IKr block with QT interval prolongation in the ECG and life-threatening cardiac arrhythmias such as TdP. Drug-induced INa and ICa block underlie the development of the BrS phenotype in experimental models of BrS.90,91

Table 4 illustrates the IC50 values for block of IKr, ICa and INa derived from heterologous expression systems (for example, HEK and CHO cells) and/or native cardiac myocytes for a number of antidepressants and antipsychotics that have been shown to induce arrhythmias.92 The available studies suggest that most antidepressants inhibit both inward and outward currents, these include imipramine, amitriptyline, and fluoxetine, all of which block both IKr and ICa. Imipramine and amitriptyline also block INa. The ability of antidepressants to block both outward and inward currents is associated with lack of correlation between the degree of IKr block and QT prolongation because calcium and/or sodium channel inhibition limit the effects of IKr block to prolong AP duration (APD) and thus to prolong the QT interval. In contrast to antidepressants, antipsychotic drugs produce more of an outward current inhibition. QT prolongation is most commonly secondary to inhibition of IKr. A 30-fold difference between the effective plasma concentration and the IC50 for inhibition of IKr has been suggested as an adequate margin of safety for avoiding the development of TdP as an adverse effect.93

Mechanisms of Arrhythmias in Long QT Syndrome

Amplification of spatial dispersion of repolarisation within the ventricular myocardium has been identified as the principal arrhythmogenic substrate in both acquired and congenital LQTS. The accentuation of spatial dispersion – typically secondary to an increase of transmural, trans-septal or apico-basal dispersion of repolarisation – and the development of early afterdepolarisation (EAD)-induced triggered activity, underlie the substrate and trigger, respectively, for the development of TdP arrhythmias observed under LQTS conditions.94,95 Models of the LQT1, LQT2, LQT3, LQT5, LQT6, LQT7, and LQT8 forms of the LQTS have been developed using the canine arterially-perfused left ventricular wedge preparations.96–99 These models suggest that in the first three forms of LQTS preferential prolongation of the M cell APD leads to an increase in the QT interval, as well as an increase in transmural dispersion of repolarisation (TDR), which contributes to the development of TdP (Figure 3).100–102 The unique characteristics of the M cells are at the heart of the LQTS. The hallmark of the M cell is the ability of its AP to prolong more than that of endocardium or epicardium in response to a slowing of rate.103–105 This feature of the M cell is a result of weaker repolarising current during phases two and three of the AP, secondary to a smaller IKs and a larger late INa and INa-Ca.106–108

Brugada Syndrome Phenotype

Article image

Antidepressant &amp; Antipsychotic Drugs

Article image

These ionic distinctions also sensitise the M cells to a variety of pharmacological agents. Agents that block IKr (such as antidepressants and antipsychotics), IKs, or increase ICa or late INa, generally produce a much greater prolongation of the APD of the M cell than that of epicardial or endocardial cells. The duration of the M cell AP therefore determines the QT interval, whereas the duration of the epicardial AP generally determines the QT peak interval.

Figure 4 presents our working hypothesis of the mechanisms underlying LQTS-related TdP. The hypothesis presumes the presence of electrical heterogeneity in the form of spatial dispersion of repolarisation in the form of transmural and trans-septal dispersion of repolarisation under baseline conditions, and the amplification of TDR by agents that reduce net repolarising current via a reduction in IKr or IKs (or augmentation of ICa or late INa). Pharmacological agents or other conditions that cause a reduction in IKr lead to a preferential prolongation of the M cell AP. As a consequence, the QT interval prolongs and is accompanied by a dramatic increase in TDR, creating a vulnerable window for the development of re-entry. The reduction in net repolarising current also predisposes to the development of EAD-induced triggered activity and, in rare cases, delayed afterdepolarisation-induced, triggered activity in M and Purkinje cells, which provide the extrasystole that triggers TdP when it falls within the vulnerable period. Betaadrenergic agonists further amplify transmural heterogeneity in the case of IKs block, as well as (transiently) in the case of IKr block, but reduce it in the case of INa agonists.102,109 Inhibition of IKr is the most common cause of reduction in net outward current by antidepressant and antipsychotic drugs. The presence of other IKr blockers (combination of an antidepressant and antipsychotic drug) or agents that reduce IKs or augment ICa or late INa can accentuate the reduction in repolarisation forces and increase the probability of arrhythmia.

Polymorphic VT Displaying Features

Article image

Arrhythmogenesis in Models of Congenital or Drug-inclued Long QT Syndrome

Article image

Mechanisms of Arrhythmia in Brugada Syndrome

The development of prominent J waves, appearing as ST-segment elevation in the right precordial leads of BrS patients, is believed to be a result of accentuation of the right ventricular (RV) epicardial AP notch secondary to an outward shift in the balance of currents active at the end of phase 1.110 A spike and dome morphology, displaying a prominent notch in ventricular epicardium but not endocardium, generates a transmural voltage gradient that results in the electrocardiographic J wave.111

The cellular basis for BrS is thought to involve an outward shift of net transmembrane current active at the end of phase 1 of the RV epicardial AP. The Ito has been shown to be most prominent in the right ventricle, particularly in the region of the RV outflow tract (RVOT).112 Such a shift can accentuate the AP notch and lead to all-or-none repolarisation at the end of phase 1 (Figures 5 and 6). When phase 1 repolarises beyond the voltage range at which L-type Ca+2 channels activate, the Ca+2 channels fail to activate, resulting in loss of the AP dome. Conduction of the AP dome from epicardial sites at which it is maintained to sites at which it is lost gives rise to phase 2 re-entry that generates a closely coupled extrasystole that precipitates VT/VF.110,113,114

Although genetic mutations are equally distributed between sexes, the clinical phenotype is 8– to10–times more prevalent in men than in women. The basis for this sex-related distinction is a more prominent Ito in the RV epicardium of men versus women.115 The more prominent Ito-mediated AP notch causes the end of phase 1 of the RV epicardial AP to repolarise to more negative potentials in tissue and arterially-perfused wedge preparations from men, facilitating loss of the AP plateau and the development of phase 2 re-entry and polymorphic VT.

The cellular mechanisms underlying BrS have long been a matter of debate.116,117 Two principal hypotheses have been proposed; the repolarisation hypothesis and the depolarisation hypothesis. The repolarisation hypothesis described above maintains that an outward shift in the balance of currents in RV epicardium can lead to repolarisation abnormalities, resulting in the development of the substrate for re-entrant activity as well as the development of phase 2 re-entry, which generates closely coupled premature beats capable of precipitating VT/VF. The depolarisation hypothesis suggests that slow conduction in the RVOT, as a result of fibrosis, reduces Cx43 expression leading to discontinuities in conduction. Conduction slowing is not necessarily limited to the RVOT area.

Leong et al. recently reported a study in which the magnitude of ST elevation correlated with the degree of ajmaline-induced conduction delay in the RVOT of patients with type I Brugada ECG, seemingly supporting the depolarisation hypothesis.118 The study included 11 patients with concealed type I BrS ECG and two healthy controls undergoing ECG imaging before and after ajmaline infusion. Activation maps and activation recovery intervals were derived from electrograms recorded from the epicardial surface of the heart, including the RV, RVOT, and left ventricle (LV). Conduction time was recorded from 3.5 cm segments within these regions of the heart before and after ajmaline and correlated with J point (ST-segment) elevation observed in the surface ECGs. Ajmaline increased conduction delay by 5.4 ± 2.8 ms in the RVOT, 2.0 ± 2.8 ms in RV free wall and 1.1 ± 1.6 ms in LV free wall. Conduction delay in the RVOT, but not RV or LV, correlated with the degree of J point elevation in BrS patients. The authors’ conclusion that the magnitude of J point (ST-segment) elevation in patients with the type I BrS pattern is attributable to conduction delay in the RVOT was challenged by Antzelevitch and Patocskai who demonstrated that ajmaline can induce prominent ST-segment elevation by accentuation of the RV epicardial AP notch.119 They further pointed out that according to the depolarisation hypothesis for ST-segment elevation associated with BrS, RVOT activation delay, relative to activation of the RV, must be roughly equivalent to the duration of the ST segment elevation (typically >200 ms)116 and that the 3.4 ms reported by Leong et al. falls far short of that requirement, thus discounting the depolarisation hypothesis as a cause.

Amitriptyline (0.2 UM) Included Brugada Phenotype

Article image

Figure 5 displays an example of amitriptyline-induced BrS phenotype in the presence of the Ito agonist NS5806 in a canine right ventricular wedge model.120 Amitriptyline (0.2 μm) caused loss of the AP dome in the AP of Epi1 but not in that of Epi2. Phase 2 re-entry developed as the epicardial AP dome propagated from sites at which it was maintained to sites at which it was lost. This mechanism generates closely coupled extrasystoles and the development of a polymorphic VT.

Genetic Predisposition

The degree to which a genetic predisposition contributes to the clinical manifestation of antidepressant- and antipsychotic-induced arrhythmogenesis is not well defined.

Congenital LQTS has been associated with 17 different genes (Table 5). Drugs are by far the most common cause of acquired forms, including drug-induced forms, of LQTS. Mounting evidence suggests that drug-induced LQTS also has a significant heritable component, and recent studies have made advances in identifying the genetic substrate underlying drug-induced LQTS. Advances in next-generation sequencing technology and molecular biology techniques have identified genetic variants underlying the acquired form of LQTS.121

Brugada Sybdrome

Article image

Acquired LQTS characterised by QT prolongation and TdP triggered by drugs, hypokalemia or bradycardia are usually reversed upon elimination of the triggers. In some cases the LQTS phenotype persists, suggesting the presence of an underlying genetic substrate.

Itoh et al. reported that a third of acquired LQTS patients carry congenital LQTS mutations, with variants in KCNH2 being the most common.122 In a recent study, Strauss et al. demonstrated that a genetic QT score comprising 61 common genetic variants can account for a significant proportion of the variability in drug-induced QT prolongation and is a significant predictor of drug-induced TdP.123 The authors indicate that these findings highlight an opportunity for such genetic discoveries to improve individualised risk–benefit assessment for pharmacologic therapies. Of note, replication of these findings in larger samples is needed to more precisely identify the individual variants that drive these effects.123

Abbott et al. were among the first to show that a polymorphism (a genetic variation that is present in greater than 1 % of the population) in an ion channel gene is associated with a predisposition to drug-induced TdP.124 They identified a polymorphism (T8A) of the KCNE2 gene encoding for MiRP, a beta subunit of the IKr channel, that is present in 1.6 % of the population and is associated with TdP related to quinidine and to sulfamethoxazole/trimethoprim administration. This finding suggests that common genetic variations may increase the risk for development of drug-related arrhythmias. Yang et al. showed that DNA variants in the coding regions of congenital long QT disease genes predisposing to acquired LQTS can be identified in approximately 10–15 % of affected subjects, predominantly in genes encoding ancillary subunits, providing further support for the hypothesis that subclinical mutations and polymorphisms may predispose to drug-induced TdP.125 Splawski et al. further advanced this concept by identifying a heterozygous polymorphism involving substitution of serine with tyrosine in codon 1103 (S1103Y) in the sodium channel gene SCN5A (S1102Y in the shorter splice variant of SCN5A) among Africans and African-Americans that increases the risk for acquired TdP.126 The polymorphism was present in 57 % of 23 patients with pro-arrhythmic episodes, but in only 13 % of controls.

Genes Associated with Congenital Long QT Syndrome

Article image

Another common polymorphism that has been associated with acquired forms of LQTS and TdP is K897T in KCNH2.127 Most functional expression studies have reported that K897T reduces IKr,128–130 although one study has reported an increase in hERG current.131

Antidepressant drugs have been shown to induce acquired LQTS via both direct inhibition of the hERG or IKr channel or via impaired trafficking of the channel to the surface membrane, thus mimicking the effects of some KCNH2 variants.132

The action of antidepressants to precipitate the BrS may also have a genetic disposition. For example, the SCN5A promoter haplotype (so-called Hap B) has been shown to be associated with longer PR and QRS intervals as well as with a more exaggerated response to sodium channel blockers.133

Genetic defects can also contribute to drug-induced channelopathies by influencing the metabolism of drugs. In the case of relatively pure IKr blockers, there is a clear relationship between plasma levels of drug and the incidence of TdP. Genetic variants of the genes encoding for enzymes responsible for drug metabolism could alter pharmacokinetics so as to cause wide fluctuations in plasma levels, thus exerting a significant proarrhythmic influence.134,135 For example, in the case of cytochrome CYP2D6, which is involved in the metabolism of some QT-prolonging drugs (terodiline, thioridazine), multiple polymorphisms have been reported that reduce or eliminate its function; 5–10 % of Caucasians and African-Americans lack a functional CYP2D6. Numerous proteins, including drug transport molecules and other drug metabolising enzymes, are involved in drug absorption, distribution and elimination, and genetic variants of each of these has the potential to modulate drug concentrations and effects. Multiple substrates and inhibitors of the cytochrome P450 enzymes have been identified. A comprehensive database can be found at http://medicine.iupui.edu/flockhart

Antipsychotic drugs are more commonly associated with QT prolongation and TdP than antidepressants are. Most cases of antidepressant-induced TdP occur following drug overdose or when administered in combination with other QT-prolonging agents or conditions. Antidepressants, on the other hand, are more likely to predispose to BrS phenotype. These proclivities are because antipsychotic drugs generally exert a predominant effect to inhibit outward currents, IKr block in particular, whereas antidepressants exert a predominant effect to inhibit inward currents, such as INa and ICa. Commonly used antipsychotic and antidepressant drugs should be used with great care in cases of long QT or BrS,or when combined with agents known to prolong QT intervals or to predispose to acquired forms of BrS.

Acknowledgements

The authors acknowledge Gan-Xin Yan, Lin Wu and Subramanian Krishnan’s assistance in providing us with ECGs of patients administered with antipsychotic and antidepressant drugs.

Clinical Perspective

  • Some antipsychotic and antidepressant drugs increase the risk of ventricular arrhythmias and SCD by prolonging the QT interval and inducing Torsade de Pointes arrhythmias. These include typical antipsychotics such as chlorpromazine; tricyclic antidepressants such as amitriptyline and other antidepressants such as fluoxetine.
  • Other antipsychotic and antidepressant drugs increase the risk of ventricular arrhythmias and SCD by inducing a Brugada Syndrome phenotype. These include antipsychotics such as trifluoperazine; tricyclic antidepressants such as amitriptyline or desipramine; and other antidepressants such as maprotiline or lithium.
  • Antipsychotic drugs can increase cardiac risk even at low doses, whereas antidepressant drugs generally do it at high doses or in combination with other drugs.
  • The newer atypical antipsychotics, including olanzapine, risperidone, quetiapine, prothipendyl, pimavanserin and benperidol display a lower level of risk than the older typical antipsychotics, especially those in the phenothiazine category.
  • Antipsychotic and antidepressant drugs should be used with great care in cases of long QT or BrS or when combined with agents known to prolong QT intervals or to predispose to acquired forms of BrS.
  • In the case of drugs categorised as having a potential to cause significant QT prolongation and/or TdP, ECG monitoring is advisable, particularly where the FDA-approved label recommends ECG monitoring. Review of specific antipsychotic or antidepressant therapy, including cessation and change of medication should be considered if the ECG shows major prolongation of the QT interval (QTc >500 ms), QTc prolongation >60 ms, T wave abnormalities, marked bradycardia, or a BrS phenotype.
  • Finally, high-risk antipsychotics and antidepressants should be avoided in patients with acute systemic disease, including acute MI and renal or hepatic disease.

References

  1. Abdelmawla N and Mitchell AJ. Sudden cardiac death and antipsychotics. Part 1: Risk factors and mechanisms. Adv Psychiatr Treat 2006;12:35–44.
    Crossref
  2. Echt DS, Liebson PR, Mitchell LB, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The Cardiac Arrhythmia Suppression Trial. N Engl J Med 1991;324:781–8.
    Crossref | PubMed
  3. Dessertenne F. [Ventricular tachycardia with 2 variable opposing foci]. Arch Mal Coeur Vaiss 1966;59:263–72 [in French].
  4. Priori SG, Schwartz PJ, Napolitano C, et al. Risk stratification in the long-QT syndrome. N Engl J Med 2003;348:1866–74.
    Crossref | PubMed
  5. Drew BJ, Ackerman MJ, Funk M, et al. Prevention of torsade de pointes in hospital settings: a scientific statement from the American Heart Association and the American College of Cardiology Foundation. J Am Coll Cardiol 2010;55:934–47.
    Crossref | PubMed
  6. Viskin S. The QT interval: too long, too short or just right. Heart Rhythm 2009;6:711–5.
    Crossref | PubMed
  7. Bednar MM, Harrigan EP, Anziano RJ, et al. The QT interval. Prog Cardiovasc Dis 2001;43:1–45.
    PubMed
  8. Tomaselli GF and Marban E. Electrophysiological remodeling in hypertrophy and heart failure. Cardiovasc Res 1999;42:270–83.
    Crossref | PubMed
  9. Sipido KR, Volders PG, De Groot SH, et al. Enhanced Ca2+ release and Na/Ca exchange activity in hypertrophied canine ventricular myocytes: potential link between contractile adaptation and arrhythmogenesis. Circulation 2000;102:2137–44.
    Crossref | PubMed
  10. Volders PG, Sipido KR, Vos MA, et al. Downregulation of delayed rectifier K(+) currents in dogs with chronic complete atrioventricular block and acquired torsades de pointes. Circulation 1999;100:2455–61.
    Crossref | PubMed
  11. Undrovinas AI, Maltsev VA and Sabbah HN. Repolarization abnormalities in cardiomyocytes of dogs with chronic heart failure: role of sustained inward current. Cell Mol Life Sci 1999;55:494–505.
    Crossref | PubMed
  12. Maltsev VA, Sabbah HN, Higgins RS, et al. Novel, ultraslow inactivating sodium current in human ventricular cardiomyocytes. Circulation 1998;98:2545–52.
    Crossref | PubMed
  13. Antzelevitch C. Role of transmural dispersion of repolarization in the genesis of drug-induced torsades de pointes. Heart Rhythm 2005;2:S9–S15.
    Crossref | PubMed
  14. Vieweg WV and Wood MA. Tricyclic antidepressants, QT interval prolongation, and torsade de pointes. Psychosomatics 2004;45:371–7.
    Crossref | PubMed
  15. Danielsson B, Collin J, Jonasdottir Bergman G, et al. Antidepressants and antipsychotics classified with torsades de pointes arrhythmia risk and mortality in older adults - a Swedish nationwide study. Br J Clin Pharmacol 2016;81:773–83.
    Crossref | PubMed
  16. Jasiak NM and Bostwick JR. Risk of QT/QTc prolongation among newer non-SSRI antidepressants. Ann Pharmacother 2014;48:1620–8.
    Crossref | PubMed
  17. Mehtonen OP, Aranko K, Malkonen L and Vapaatalo H. A survey of sudden death associated with the use of antipsychotic or antidepressant drugs: 49 cases in Finland. Acta Psychiatr Scand. 1991;84:58–64.
    Crossref | PubMed
  18. Ray WA, Meredith S, Thapa PB, et al. Antipsychotics and the risk of sudden cardiac death. Arch Gen Psychiatry 2001;58:1161–7.
    Crossref | PubMed
  19. Hennessy S, Bilker WB, Knauss JS, et al. Cardiac arrest and ventricular arrhythmia in patients taking antipsychotic drugs: cohort study using administrative data. BMJ 2002;325:1070.
    Crossref | PubMed
  20. Liperoti R, Gambassi G, Lapane KL, et al. Conventional and atypical antipsychotics and the risk of hospitalization for ventricular arrhythmias or cardiac arrest. Arch Intern Med 2005;165:696–701.
    Crossref | PubMed
  21. Ozeki Y, Fujii K, Kurimoto N, et al. QTc prolongation and antipsychotic medications in a sample of 1017 patients with schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2010;34:401–5.
    Crossref | PubMed
  22. Beach SR, Celano CM, Noseworthy PA, et al. QTc prolongation, torsades de pointes, and psychotropic medications. Psychosomatics 2013;54:1–13.
    Crossref | PubMed
  23. Brugada P and Brugada J. Right bundle branch block, persistent ST segment elevation and sudden cardiac death: a distinct clinical and electrocardiographic syndrome: a multicenter report. J Am Coll Cardiol 1992;20:1391–6.
    Crossref | PubMed
  24. Wilde AA, Antzelevitch C, Borggrefe M, et al. Proposed diagnostic criteria for the Brugada syndrome. Eur Heart J 2002;23:1648–54.
    Crossref | PubMed
  25. Wilde AA, Antzelevitch C, Borggrefe M, et al. Proposed diagnostic criteria for the Brugada syndrome: consensus report. Circulation 2002;106:2514–9.
    Crossref | PubMed
  26. Antzelevitch C, Brugada P, Borggrefe M, et al. Brugada syndrome: report of the second consensus conference: endorsed by the Heart Rhythm Society and the European Heart Rhythm Association. Circulation 2005;111:659–70.
    Crossref | PubMed
  27. Antzelevitch C, Brugada P, Borggrefe M, et al. Brugada syndrome: report of the second consensus conference. Heart Rhythm 2005;2:429–40.
    Crossref | PubMed
  28. Antzelevitch C, Yan GX, Ackerman MJ, et al. J-Wave syndromes expert consensus conference report: Emerging concepts and gaps in knowledge. Heart Rhythm 2016;13:e295–324.
    Crossref | PubMed
  29. Probst V, Veltmann C, Eckardt L, et al. Long-term prognosis of patients diagnosed with Brugada syndrome: results from the FINGER Brugada Syndrome Registry. Circulation 2010;121:635–43.
    Crossref | PubMed
  30. Kamakura S, Ohe T, Nakazawa K, et al. Long-term prognosis of probands with Brugada-pattern ST-elevation in leads V1-V3. Circ Arrhythm Electrophysiol 2009;2:495–503.
    Crossref | PubMed
  31. Conte G, Sieira J, Ciconte G, et al. Implantable cardioverter-defibrillator therapy in brugada syndrome: a 20-year single-center experience. J Am Coll Cardiol 2015;65:879–88.
    Crossref | PubMed
  32. Priori SG, Napolitano C, Glordano U, et al. Brugada syndrome and sudden cardiac death in children. Lancet 2000;355:808–9.
    Crossref | PubMed
  33. Probst V, Denjoy I, Meregalli PG, et al. Clinical aspects and prognosis of Brugada syndrome in children. Circulation 2007;115:2042–8.
    Crossref | PubMed
  34. Probst V, Veltmann C, Eckardt L, et al. Long-term prognosis of patients diagnosed with Brugada syndrome: Results from the FINGER Brugada Syndrome Registry. Circulation 2010;121:635–43.
    Crossref | PubMed
  35. Benito B, Sarkozy A, Mont L, et al. Gender differences in clinical manifestations of Brugada syndrome. J Am Coll Cardiol 2008;52:1567–1573.
    Crossref | PubMed
  36. Sieira J, Conte G, Ciconte G, et al. Clinical characterisation and long-term prognosis of women with Brugada syndrome. Heart 2016;102:452–8.
    Crossref | PubMed
  37. Antzelevitch C. J wave syndromes: molecular and cellular mechanisms. J Electrocardiol 2013;46:510–8.
    Crossref | PubMed
  38. Antzelevitch C. Genetic, molecular and cellular mechanisms underlying the J wave syndromes. Circ J 2012;76:1054–65.
    Crossref | PubMed
  39. Kapplinger JD, Tester DJ, Alders M, et al. An international compendium of mutations in the SCN5A encoded cardiac sodium channel in patients referred for Brugada syndrome genetic testing. Heart Rhythm 2010;7:33–46.
    Crossref | PubMed
  40. Ackerman MJ, Priori SG, Willems S, et al. HRS/EHRA expert consensus statement on the state of genetic testing for the channelopathies and cardiomyopathies this document was developed as a partnership between the Heart Rhythm Society (HRS) and the European Heart Rhythm Association (EHRA). Heart Rhythm 2011;8:1308–39.
    Crossref | PubMed
  41. Burashnikov E, Pfeiffer R, Barajas-Martinez H, et al. Mutations in the cardiac L-type calcium channel associated J wave syndrome and sudden cardiac death. Heart Rhythm 2010;7:1872–82.
    Crossref | PubMed
  42. Cordeiro JM, Marieb M, Pfeiffer R, et al. Accelerated inactivation of the L-type calcium due to a mutation in CACNB2b due to a mutation in CACNB2b underlies Brugada syndrome. J Mol Cell Cardiol 2009;46:695–703.
    Crossref | PubMed
  43. Antzelevitch C, Pollevick GD, Cordeiro JM, et al. Loss-of-function mutations in the cardiac calcium channel underlie a new clinical entity characterized by ST-segment elevation, short QT intervals, and sudden cardiac death. Circulation 2007;115(4):442-9.
    Crossref | PubMed
  44. Gurnett CA, De WM and Campbell KP. Dual function of the voltage-dependent Ca2+ channel alpha 2 delta subunit in current stimulation and subunit interaction. Neuron 1996;16:431–40.
    Crossref | PubMed
  45. Barajas-Martinez H, Hu D, Ferrer T, et al. Molecular genetic and functional association of Bugada and early repolarization syndromes with S422L missense mutation in KCNJ8. Heart Rhythm 2012;9:548–55.
    Crossref | PubMed
  46. Delaney JT, Muhammad R, Blair MA, et al. A KCNJ8 mutation associated with early repolarization and atrial fibrillation. Europace 2012;14:1428–32.
    Crossref | PubMed
  47. Medeiros-Domingo A, Tan BH, Crotti L, et al. Gain-of-function mutation S422L in the KCNJ8-encoded cardiac K(ATP) channel Kir6.1 as a pathogenic substrate for J-wave syndromes. Heart Rhythm 2010;7:1466–71.
    Crossref | PubMed
  48. Hu D, Barajas-Martinez H, Medeiros-Domingo A, et al. Novel mutations in the sodium channel 2 subunit gene (SCN2B) associated with Brugada syndrome and atrial fibrillation. Circulation 2012;126:A16521.
  49. Riuro H, Beltran-Alvarez P, Tarradas A, et al. A missense mutation in the sodium channel ß2 subunit reveals SCN2B as a new candidate gene for Brugada syndrome. Hum Mutat 2013;34:961–6.
    Crossref | PubMed
  50. Giudicessi JR, Ye D, Tester DJ, et al. Transient outward current (Ito) gain-of-function mutations in the KCND3-encoded Kv4.3 potassium channel and Brugada syndrome. Heart Rhythm 2011;8:1024–32.
    Crossref | PubMed
  51. Delpón E, Cordeiro JM, Núñez L, et al. Functional effects of KCNE3 mutation and its role in the development of Brugada syndrome. Circ Arrhythm Electrophysiol 2008;1:209–18.
    Crossref | PubMed
  52. Olesen MS, Jensen NF, Holst AG, et al. A novel nonsense variant in Nav1.5 cofactor MOG1 eliminates its sodium current increasing effect and may increase the risk of arrhythmias. Can J Cardiol 2011;27:523.
    Crossref | PubMed
  53. Kattygnarath D, Maugenre S, Neyroud N, et al. MOG1: a new susceptibility gene for Brugada syndrome. Circ Cardiovasc Genet 2011;4:261–8.
    Crossref | PubMed
  54. Cerrone M, Lin X, Zhang M, et al. Missense mutations in plakophilin-2 cause sodium current deficit and associate with a Brugada syndrome phenotype. Circulation 2014;129:1092–03.
    Crossref | PubMed
  55. Hennessey JA, Marcou CA, Wang C, et al. FGF12 is a candidate Brugada syndrome locus. Heart Rhythm 2013;10:1886–94.
    Crossref | PubMed
  56. Bezzina CR, Barc J, Mizusawa Y, et al. Common variants at SCN5A-SCN10A and HEY2 are associated with Brugada syndrome, a rare disease with high risk of sudden cardiac death. Nat Genet 2013;45:1044–9.
    Crossref | PubMed
  57. Hu D, Barajas-Martinez H, Pfeiffer R, et al. Mutations in SCN10A are responsible for a large fraction of cases of Brugada syndrome. J Am Coll Cardiol 2014;64:66–79.
    Crossref | PubMed
  58. Behr ER, Savio-Galimberti E, Barc J, et al. Role of common and rare variants in SCN10A: results from the Brugada syndrome QRS locus gene discovery collaborative study. Cardiovasc Res 2015;106:520–9.
    Crossref | PubMed
  59. Antzelevitch C. Cardiac repolarization. The long and short of it. Europace 2005;7(Suppl 2):3–9.
    Crossref | PubMed
  60. Perrin MJ, Adler A, Green S, et al. Evaluation of genes encoding for the transient outward current (Ito) identifies the KCND2 gene as a cause of J wave syndrome associated with sudden cardiac death. Circ Cardiovasc Genet 2014; 7:782–9.
    Crossref | PubMed
  61. Liu H, Chatel S, Simard C, et al. Molecular genetics and functional anomalies in a series of 248 Brugada cases with 11 mutations in the TRPM4 channel. PLoS ONE 2013;8:e54131.
    Crossref | PubMed
  62. Verkerk AO, Wilders R, Schulze-Bahr E, et al. Role of sequence variations in the human ether-a-go-go-related gene (HERG, KCNH2) in the Brugada syndrome 1. Cardiovasc Res 2005;68:441–53.
    Crossref | PubMed
  63. Wilders R and Verkerk AO. Role of the R1135H KCNH2 mutation in Brugada syndrome. IntJ Cardiol 2010;144:149–51.
    Crossref | PubMed
  64. Ohno S, Zankov DP, Ding WG, et al. KCNE5 (KCNE1L) variants are novel modulators of Brugada syndrome and idiopathic ventricular fibrillation. Circ Arrhythm Electrophysiol 2011;4:352–61.
    Crossref | PubMed
  65. Boczek NJ, Ye D, Johnson EK, et al. Characterization of SEMA3A-encoded semaphorin as a naturally occurring Kv4.3 protein inhibitor and its contribution to Brugada syndrome. Circ Res 2014;115:460–9;
    Crossref | PubMed
  66. Ueda K, Nakamura K, Hayashi T, et al. Functional characterization of a trafficking-defective HCN4 mutation, D553N, associated with cardiac arrhythmia. J Biol Chem 2004;279:27194–8.
    Crossref | PubMed
  67. Brugada P, Brugada J and Brugada R. Arrhythmia induction by antiarrhythmic drugs. Pacing Clin Electrophysiol 2000;23:291–2.
    Crossref | PubMed
  68. Brugada R, Brugada J, Antzelevitch C, et al. Sodium channel blockers identify risk for sudden death in patients with ST-segment elevation and right bundle branch block but structurally normal hearts. Circulation 2000;101:510–5.
    Crossref | PubMed
  69. Miyazaki T, Mitamura H, Miyoshi S, et al. Autonomic and antiarrhythmic drug modulation of ST segment elevation in patients with Brugada syndrome. J Am Coll Cardiol 1996;27:1061–70.
    Crossref | PubMed
  70. Babaliaros VC and Hurst JW. Tricyclic antidepressants and the Brugada syndrome: an example of Brugada waves appearing after the administration of desipramine. Clin Cardiol 2002;25:395–8.
    Crossref | PubMed
  71. Goldgran-Toledano D, Sideris G and Kevorkian JP. Overdose of cyclic antidepressants and the Brugada syndrome. N Engl J Med 2002;346:1591–2.
    Crossref | PubMed
  72. Tada H, Sticherling C, Oral H and Morady F. Brugada syndrome mimicked by tricyclic antidepressant overdose. J Cardiovasc Electrophysiol 2001;12:275.
    Crossref | PubMed
  73. Pastor A, Nunez A, Cantale C and Cosio FG. Asymptomatic Brugada syndrome case unmasked during dimenhydrinate infusion. J Cardiovasc Electrophysiol 2001;12:1192–4.
    Crossref | PubMed
  74. Ortega-Carnicer J, Bertos-Polo J and Gutierrez-Tirado C. Aborted sudden death, transient Brugada pattern, and wide QRS dysrrhythmias after massive cocaine ingestion. J Electrocardiol 2001;34:345–9.
    Crossref | PubMed
  75. Nogami A, Nakao M, Kubota S, et al. Enhancement of J-ST-segment elevation by the glucose and insulin test in Brugada syndrome. Pacing Clin Electrophysiol 2003;26:332–7.
    Crossref | PubMed
  76. Araki T, Konno T, Itoh H, et al. Brugada syndrome with ventricular tachycardia and fibrillation related to hypokalemia. Circ J 2003;67:93–5.
    Crossref | PubMed
  77. Akhtar M and Goldschlager NF. Brugada electrocardiographic pattern due to tricyclic antidepressant overdose. J Electrocardiol 2006;39:336–9.
    Crossref | PubMed
  78. Matana A, Goldner V, Stanic K, et al. Unmasking effect of propafenone on the concealed form of the Brugada phenomenon. Pacing Clin Electrophysiol 2000;23:416–8.
    Crossref | PubMed
  79. Hasdemir C, Olukman M, Ulucan C and Roden DM. Brugada-type ECG pattern and extreme QRS complex widening with propafenone overdose. J Cardiovasc Electrophysiol 2006;17:565–6.
    Crossref | PubMed
  80. Fragakis N, Iliadis I, Papanastasiou S, et al. Brugada type electrocardiographic changes induced by concomitant use of lithium and propafenone in patient with wolff-Parkinson-white syndrome. Pacing Clin Electrophysiol 2007;30:823–5.
    Crossref | PubMed
  81. Chutani S, Imran N, Grubb B and Kanjwal Y. Propafenone-induced Brugada-like ECG changes mistaken as acute myocardial infarction. Emerg Med J 2008;25:117–8.
    Crossref | PubMed
  82. Darbar D, Yang T, Churchwell K, et al. Unmasking of Brugada syndrome by lithium. Circulation 2005;112:1527–31.
    Crossref | PubMed
  83. Riera AR, Uchida AH, Schapachnik E, et al. Propofol infusion syndrome and Brugada syndrome electrocardiographic phenocopy. Cardiol J 2010;17:130–5.
    PubMed
  84. Vernooy K, Delhaas T, Cremer OL, et al. Electrocardiographic changes predicting sudden death in propofol-related infusion syndrome. Heart Rhythm 2006;3:131–7.
    Crossref | PubMed
  85. Rouleau F, Asfar P, Boulet S, et al. Transient ST segment elevation in right precordial leads induced by psychotropic drugs: relationship to the Brugada syndrome. J Cardiovasc Electrophysiol 2001;12:61–5.
    Crossref | PubMed
  86. Chow BJ, Gollob M and Birnie D. Brugada syndrome precipitated by a tricyclic antidepressant. Heart 2005;91:651.
    Crossref | PubMed
  87. Bolognesi R, Tsialtas D, Vasini P, et al. Abnormal ventricular repolarization mimicking myocardial infarction after heterocyclic antidepressant overdose. Am J Cardiol 1997;79:242–5.
    Crossref | PubMed
  88. Bigwood B, Galler D, Amir N and Smith W. Brugada syndrome following tricyclic antidepressant overdose. Anaesth Intensive Care 2005;33:266–70.
    PubMed
  89. Pirotte MJ, Mueller JG and Poprawski T. A case report of Brugada-type electrocardiographic changes in a patient taking lithium. Am J Emerg Med 2008;26:113.
    Crossref | PubMed
  90. Fish JM and Antzelevitch C. Role of sodium and calcium channel block in unmasking the Brugada syndrome. Heart Rhythm 2004;1:210–7.
    Crossref | PubMed
  91. Antzelevitch C and Fish JM. Therapy for the Brugada syndrome. Handb Exp Pharmacol 2006:305–30.
    PubMed
  92. Sala M, Coppa F, Cappucciati C, et al. Antidepressants: their effects on cardiac channels, QT prolongation and Torsade de Pointes. Curr Opin Investig Drugs 2006;7:256–63.
    PubMed
  93. Redfern WS, Carlsson L, Davis AS, et al. Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development. Cardiovasc Res 2003;58:32–45.
    Crossref | PubMed
  94. Belardinelli L, Antzelevitch C and Vos MA. Assessing predictors of drug-induced torsade de pointes. Trends Pharmacol Sci 2003;24:619–25.
    Crossref | PubMed
  95. Antzelevitch C and Shimizu W. Cellular mechanisms underlying the long QT syndrome. Curr Opin Cardiol 2002;17:43–51.
    Crossref | PubMed
  96. Shimizu W and Antzelevitch C. Effects of a K+ channel opener to reduce transmural dispersion of repolarization and prevent torsade de pointes in LQT1, LQT2, and LQT3 models of the long-QT syndrome. Circulation 2000;102:706–12.
    Crossref | PubMed
  97. Antzelevitch C. Heterogeneity of cellular repolarization in LQTS: the role of M cells. Eur Heart J Suppl 2001;3:K2–K16.
  98. Tsuboi M and Antzelevitch C. Cellular basis for electrocardiographic and arrhythmic manifestations of Andersen-Tawil syndrome (LQT7). Heart Rhythm 2006;3:328–35.
    Crossref | PubMed
  99. Sicouri S, Timothy KW, Zygmunt AC, et al. Cellular basis for the electrocardiographic and arrhythmic manifestations of Timothy syndrome: effects of ranolazine. Heart Rhythm 2007;4:638–47.
    Crossref | PubMed
  100. Shimizu W and Antzelevitch C. Cellular basis for the ECG features of the LQT1 form of the long QT syndrome: effects of beta−adrenergic agonists and antagonists and sodium channel blockers on transmural dispersion of repolarization and torsade de pointes. Circulation 1998;98:2314–22.
    Crossref | PubMed
  101. Shimizu W and Antzelevitch C. Sodium channel block with mexiletine is effective in reducing dispersion of repolarization and preventing torsade de pointes in LQT2 and LQT3 models of the long-QT syndrome. Circulation 1997;96:2038–47.
    Crossref | PubMed
  102. Shimizu W and Antzelevitch C. Differential effects of beta-adrenergic agonists and antagonists in LQT1, LQT2 and LQT3 models of the long QT syndrome. J Am Coll Cardiol 2000;35:778–86.
    Crossref | PubMed
  103. Sicouri S and Antzelevitch C. A subpopulation of cells with unique electrophysiological properties in the deep subepicardium of the canine ventricle. The M cell. Circ Res 1991;68:1729–41.
    Crossref | PubMed
  104. Antzelevitch C, Shimizu W, Yan GX, et al. The M cell: its contribution to the ECG and to normal and abnormal electrical function of the heart. J Cardiovasc Electrophysiol 1999;10:1124–52.
    Crossref | PubMed
  105. Anyukhovsky EP, Sosunov EA, Gainullin RZ and Rosen MR. The controversial M cell. J Cardiovasc Electrophysiol 1999;10:244–60.
    Crossref | PubMed
  106. Zygmunt AC, Goodrow RJ and Antzelevitch C. INaCa contributes to electrical heterogeneity within the canine ventricle. Am J Physiol Heart Circ Physiol 2000;278:H1671-8.
    Crossref | PubMed
  107. Zygmunt AC, Eddlestone GT, Thomas GP, et al. Larger late sodium conductance in M cells contributes to electrical heterogeneity in canine ventricle. Am J Physiol 2001;281:H689–97.
    Crossref | PubMed
  108. Liu DW and Antzelevitch C. Characteristics of the delayed rectifier current (IKr and IKs) in canine ventricular epicardial, midmyocardial, and endocardial myocytes. A weaker IKs contributes to the longer action potential of the M cell. Circ Res 1995;76:351–65.
    Crossref | PubMed
  109. Li GR, Feng J, Yue L and Carrier M. Transmural heterogeneity of action potentials and Ito1 in myocytes isolated from the human right ventricle. Am J Physiol 1998;275:H369–77.
    Crossref | PubMed
  110. Antzelevitch C. The Brugada syndrome: diagnostic criteria and cellular mechanisms. Eur Heart J 2001;22:356–63.
    Crossref | PubMed
  111. Yan GX and Antzelevitch C. Cellular basis for the electrocardiographic J wave. Circulation 1996;93:372–9.
    Crossref | PubMed
  112. Volders PG, Sipido KR, Carmeliet E, et al. Repolarizing K+ currents ITO1 and IKs are larger in right than left canine ventricular midmyocardium. Circulation 1999;99:206–10.
    Crossref | PubMed
  113. Antzelevitch C. State of the art: overview of brugada syndrome. Circulation Journal 2006;70:12.
  114. Badri M, Patel A and Yan G. Cellular and ionic basis of J-wave syndromes. Trends Cardiovasc Med 2015;25:12–21.
    Crossref | PubMed
  115. Di Diego JM, Cordeiro JM, Goodrow RJ, et al. Ionic and cellular basis for the predominance of the Brugada syndrome phenotype in males. Circulation 2002;106:2004–11.
    Crossref | PubMed
  116. Wilde AA, Postema PG, Di Diego JM, et al. The pathophysiological mechanism underlying Brugada syndrome: depolarization versus repolarization. J Mol Cell Cardiol 2010;49:543–53.
    Crossref | PubMed
  117. Morita H, Zipes DP and Wu J. Brugada syndrome: insights of ST elevation, arrhythmogenicity, and risk stratification from experimental observations. Heart Rhythm 2009;6:S34–43.
    Crossref | PubMed
  118. Leong KM, Ng FS, Yao C, et al. ST-Elevation Magnitude Correlates With Right Ventricular Outflow Tract Conduction Delay in Type I Brugada ECG. Circ Arrhythm Electrophysiol 2017;10:e005107.
    Crossref | PubMed
  119. Antzelevitch C and Patocskai B. Ajmaline-Induced Slowing of Conduction in the Right Ventricular Outflow Tract Cannot Account for ST Elevation in Patients With Type I Brugada ECG. Circ Arrhythm Electrophysiol 2017;10:e005775.
    Crossref | PubMed
  120. Minoura Y, Di Diego JM, Barajas-Martinez H, et al. Ionic and cellular mechanisms underlying the development of acquired Brugada syndrome in patients treated with antidepressants. J Cardiovasc Electrophysiol 2012;23:423–32.
    Crossref | PubMed
  121. Mahida S, Hogarth AJ, Cowan C, et al. Genetics of congenital and drug-induced long QT syndromes: current evidence and future research perspectives. J Interv Card Electrophysiol 2013;37:9–19.
    Crossref | PubMed
  122. Itoh H, Crotti L, Aiba T, et al. The genetics underlying acquired long QT syndrome: impact for genetic screening. Eur Heart J 2016;37:1456–64.
    Crossref | PubMed
  123. Strauss DG, Vicente J, Johannesen L, et al. Common Genetic Variant Risk Score Is Associated With Drug-Induced QT Prolongation and Torsade de Pointes Risk: A Pilot Study. Circulation 2017;135:1300–10.
    Crossref | PubMed
  124. Abbott GW, Sesti F, Splawski I, et al. MiRP1 forms IKr potassium channels with HERG and is associated with cardiac arrhythmia. Cell 1999;97:175–87.
    Crossref | PubMed
  125. Yang P, Kanki H, Drolet B, et al. Allelic variants in long-QT disease genes in patients with drug-associated torsades de pointes. Circulation 2002;105:1943–8.
    Crossref | PubMed
  126. Splawski I, Timothy KW, Tateyama M, et al. Variant of SCN5A sodium channel implicated in risk of cardiac arrhythmia. Science 2002;297:1333–6.
    Crossref | PubMed
  127. Pollevick GD, Oliva A, Viskin S, et al. Genetic predisposition to post-myocardial infarction long QT intervals and torsade de pointes. Heart Rhythm 2007;4:S121.
  128. Crotti L, Lundquist AL, Insolia R, et al. KCNH2-K897T is a genetic modifier of latent congenital long-QT syndrome. Circulation 2005;112:1251–8.
    Crossref | PubMed
  129. Anson BD, Ackerman MJ, Tester DJ, et al. Molecular and functional characterization of common polymorphisms in HERG (KCNH2) potassium channels. Am J Physiol Heart Circ Physiol 2004;286:H2434–41.
    Crossref | PubMed
  130. Paavonen KJ, Chapman H, Laitinen PJ, et al. Functional characterization of the common amino acid 897 polymorphism of the cardiac potassium channel KCNH2 (HERG). Cardiovasc Res 2003;59:603–11.
    Crossref | PubMed
  131. Bezzina CR, Verkerk AO, Busjahn A, et al. A common polymorphism in KCNH2 (HERG) hastens cardiac repolarization. Cardiovasc Res 2003;59:27–36.
    Crossref | PubMed
  132. Cubeddu LX. Drug-induced Inhibition and Trafficking Disruption of ion Channels: Pathogenesis of QT Abnormalities and Drug-induced Fatal Arrhythmias. Curr Cardiol Rev 2016;12:141–54.
    Crossref | PubMed
  133. Bezzina CR, Shimizu W, Yang P, et al. Common sodium channel promoter haplotype in asian subjects underlies variability in cardiac conduction. Circulation 2006;113:338–44.
    Crossref | PubMed
  134. Ford GA, Wood SM and Daly AK. CYP2D6 and CYP2C19 genotypes of patients with terodiline cardiotoxicity identified through the yellow card system. BrJ Clin Pharmacol 2000;50:77–80.
    Crossref | PubMed
  135. Roden DM. Pharmacogenetics and drug-induced arrhythmias. Cardiovasc Res 2001;50:224–31.
    Crossref | PubMed
  136. Dorsey ST and Biblo LA. Prolonged QT interval and torsades de pointes caused by the combination of fluconazole and amitriptyline. Am J Emerg Med 2000;18:227–9.
    Crossref | PubMed
  137. Davison ET. Amitriptyline-induced Torsade de Pointes. Successful therapy with atrial pacing. J Electrocardiol 1985;18:299–301.
    Crossref | PubMed
  138. Jerjes Sánchez Díaz C, García Hernández N, González Carmona VM, Rosado Buzzo A. [Helicoidal ventricular tachycardia caused by amitriptyline. Presentation of a case]. Arch Inst Cardiol Mex 1985;55:353–6 [in Spanish].
    PubMed
  139. Flugelman MY, Pollack S, Hammerman H, et al. Congenital prolongation of Q-T interval: a family study of three generations. Cardiology 1982;69:170–4.
    Crossref | PubMed
  140. Strasberg B, Coelho A, Welch W, et al. Doxepin induced torsade de pointes. Pacing Clin Electrophysiol. 1982;5:873–7.
    Crossref | PubMed
  141. Alter P, Tontsch D and Grimm W. Doxepin-induced torsade de pointes tachycardia. Ann Intern Med 2001;135:384–5.
    Crossref | PubMed
  142. Stollberger C, Huber JO and Finsterer J. Antipsychotic drugs and QT prolongation. Int Clin Psychopharmacol 2005;20:243–51.
    Crossref | PubMed
  143. Alampay MM, Haigney MC, Flanagan MC, et al. Transcranial magnetic stimulation as an antidepressant alternative in a patient with Brugada syndrome and recurrent syncope. Mayo Clin Proc 2014;89:1584–7.
    Crossref | PubMed
  144. Yap YG and Camm AJ. Drug induced QT prolongation and torsades de pointes. Heart. 2003;89:1363–72.
    Crossref | PubMed
  145. Teschemacher AG, Seward EP, Hancox JC and Witchel HJ. Inhibition of the current of heterologously expressed HERG potassium channels by imipramine and amitriptyline. Br J Pharmacol 1999;128:479–85.
    Crossref | PubMed
  146. Jo SH, Youm JB, Lee CO, et al. Blockade of the HERG human cardiac K(+) channel by the antidepressant drug amitriptyline. Br J Pharmacol 2000;129:1474–80.
    Crossref | PubMed
  147. Park KS, Kong ID, Park KC and Lee JW. Fluoxetine inhibits L-type Ca2+ and transient outward K+ currents in rat ventricular myocytes. Yonsei Med J 1999;40:144–51.
    Crossref | PubMed
  148. Barber MJ, Starmer CF and Grant AO. Blockade of cardiac sodium channels by amitriptyline and diphenylhydantoin. Evidence for two use-dependent binding sites. Circ Res 1991;69:677–96.
    Crossref | PubMed
  149. Curtis LH, Ostbye T, Sendersky V, et al. Prescription of QT-prolonging drugs in a cohort of about 5 million outpatients. Am J Med 2003;114:135–41.
    Crossref | PubMed
  150. Witchel HJ, Hancox JC and Nutt DJ. Psychotropic drugs, cardiac arrhythmia, and sudden death. J Clin Psychopharmacol 2003;23:58–77.
    Crossref | PubMed
  151. Thomas D, Gut B, Wendt-Nordahl G and Kiehn J. The antidepressant drug fluoxetine is an inhibitor of human ether-a-go-go-related gene (HERG) potassium channels. J Pharmacol Exp Ther 2002;300:543–8.
    Crossref | PubMed
  152. Tie H, Walker BD, Valenzuela SM, et al. The heart of psychotropic drug therapy. Lancet 2000;355:1825.
    Crossref | PubMed
  153. Tie H, Walker BD, Singleton CB, et al. Clozapine and sudden death. J Clin Psychopharmacol 2001;21:630–2.
    Crossref | PubMed
  154. Tamargo J. Drug-induced torsade de pointes: from molecular biology to bedside. Jpn J Pharmacol 2000;83:1–19.
    Crossref | PubMed
  155. Obeyesekere MN, Antzelevitch C and Krahn AD. Management of ventricular arrhythmias in suspected channelopathies. Circ Arrhythm Electrophysiol 2015;8:221–31.
    Crossref | PubMed