Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2014 Mar 20;77(4):618–625. doi: 10.1111/bcp.12208

The genetics of pro-arrhythmic adverse drug reactions

Evmorfia Petropoulou 1, Yalda Jamshidi 1, Elijah R Behr 2
PMCID: PMC3971979  PMID: 23834499

Abstract

Ventricular arrhythmia induced by drugs (pro-arrythmia) is an uncommon event, whose occurrence is unpredictable but potentially fatal. The ability of a variety of medications to induce these arrhythmias is a significant problem facing the pharmaceutical industry. Genetic variants have been shown to play a role in adverse events and are also known to influence an individual's optimal drug dose. This review provides an overview of the current understanding of the role of genetic variants in modulating the risk of drug induced arrhythmias.

Keywords: adverse drug reactions, genetics, genomics, long QT syndrome, pro-arrhythmia, QT interval

Does pro-arrhythmia have a genetic predisposition?

Drug-induced arrhythmia, also known as pro-arrhythmia, can be identified in up to 5% of patients receiving anti-arrhythmic drugs such as amiodarone, sotalol, flecainide and quinidine as well as a much smaller proportion who take non-cardiovascular medications such as antidepressants, antipsychotics, antibiotics and methadone (Table 1) 1. Clinical risk factors have been identified for drug-induced arrhythmia, and these include gender, pre-existent cardiac disease, bradyarrhythmias and liver disease (Table 2) 25.

Table 1.

Prominent examples of drugs that have been found to be associated with drug-induced long QT syndrome (adapted by: Behr & Roden 5; Ramirez et al. 30)

Drug group Drugs associated with drug-induced arrhythmias
Anti-arrhythmics class I Quinidine, procainamide, disopyramide, dihydroquinidine, bretyllium
Anti-arrhythmics class III Sotalol, dofetilide, azimilide, Ibutilide, almokalant
Anti-anginals and vasodilators Prenylamine, terodiline, lidoflazine, bepridil
Antihypertensives Nicardipine, isradipine
Antihistamines Terfenadine, astemizole
Serotonin agonists and antagonists Cisapride, ketanserin
Antimicrobials
 Macrolides Erythromycin, spiramycin, azithromycin
 Antivirals Ganciclovir, foscarnet
 Fluoroquinolones Moxifloxacin, ciprofloxacin
 Antifungals Ketoconazole, fluconazole, voriconazole
 Antimalarials Halofantrine, quinine sulphate, chloroquine
Psychiatric medications
 Tricyclic antidepressants Amitriptyline, desipramine, imipramine, doxepin,
 Antipsychotics Thioridazine, chlorpromazine, haloperidol, droperidol
 Serotonin re-uptake inhibitors Citalopram, fluoxetine, sertraline, venlafaxine
Anti-emetic and gut motility Metoclopramide, domperidone
Anticancer Arsenic trioxide, vandetanibe, eribulin
Others Probucol, methadone, pentamidine, sirolimus, lithium

Table 2.

Clinical risk factors that have been identified for drug-induced arrhythmias

Demographic factors
 Female gender
 Age (over 65 years)
Cardiac factors
 Myocardial hypertrophy
 Heart failure
 Conversion of atrial fibrillation to sinus rhythm
 Slow heart rhythms (bradycardia)
 Undiagnosed congenital long QT syndrome
Pharmacological factors
 Increased medication bioavailability
 Inhibitory medications
Others
 Electrolyte imbalance – hypokalaemia, hypomagnesaemia
 Liver disease
 Altered function of particular cytochrome P450 (CYP450) isoforms

Drug-induced arrhythmia is predominantly related to torsades de pointes (TdP) and is known as drug-induced long QT syndrome (DILQTS) or drug-induced TdP (DITdP). TdP is a characteristic form of polymorphic ventricular tachycardia that can lead to syncope and sudden cardiac death and is strongly associated with QT interval prolongation 5,6. The QT interval (Figure 1) measures the electrical depolarization, repolarization and plateau phases of the left and right ventricles of the heart on the electrocardiogram (ECG). QT prolongation is defined as the QTc interval being greater than 440 ms in men and 460 ms in women, although arrhythmias are mainly associated with a QTc of >500 ms. In the general population the change in the QT interval is associated with risk for sudden cardiac death 710. DITdP is similar to TdP seen in congenital LQTS (cLQTS) which is a result of mutations that disrupt cardiac sodium (Na+), calcium (Ca2+) and potassium (K+) ion channel function, such that repolarization of the heart is delayed 11.

Figure 1.

Figure 1

The figure demonstrates an ECG rhythm strip of a heartbeat in which the QT interval is normal in contrast with an abnormal heartbeat in which the QT interval is prolonged. This is correlated with the ventricular and Purkinje myocyte action potential with associated prolongation of phases 2 and 3. The hashed line represents the prolonged QT interval

The depolarization phase of the heart depends mainly on the inward current movement of the Na+ channel, the repolarization phase of the heart depends on the outward current movement of the K+ channel and the plateau phase depends on the balance between the repolarizing outward K+ currents and depolarizing L-type inward currents of the Ca2+ channel. Anti-arrhythmic and non-anti-arrhythmic drugs have been demonstrated to block these ion channels leading to diminished currents which can sometimes result in drug-induced adverse reactions. These pro-arrhythmic adverse reactions are likely to be generated due to an association between the patient-specific and drug-specific clinical risk factors 12. Two such factors associated with patient-specific risk include: (i) pharmacokinetic factors which are responsible for regulating the drug concentration transferred to drug receptors and (ii) pharmacodynamic factors which control the response to doses of drugs 13.

One of the basic mechanisms playing an essential role in the repolarization of the human heart is the time-dependent outward rectifying K+ current (IK) 14. Several studies since 1980 have demonstrated that this current has many subtypes of which the most important are: IKr and IKs (‘r’ stands for rapid and ‘s’ stands for slow) 15,16. Mutations that lead to loss of function in genes underlying even one of these two currents may be responsible for cLQTS. The inhibition of IKr is the main mechanism by which prolongation of the QT interval is caused by almost all culpable drugs 17. Pro-arrhythmia resulting from cardiovascular and/or non-cardiovascular drugs could therefore be a result of the inhibition of IKr with or without diminished IK due to mutations in the underlying genes. This was conceptualized as the repolarization reserve by Roden who stated that ‘the complexity of repolarization includes some redundancy’ 18. Thus loss of one ionic current subtype such as IKr, will not necessarily result in complete failure of repolarization. As a result individuals with subclinical genetic lesions in different ionic currents such as IKs or even other ion currents such as Na+ or Ca2+, might not demonstrate any overt change in repolarization as suggested by Xiao et al. 19.

Strong evidence for a genetic basis to drug-induced arrhythmia was provided by Kannankeril et al. 20. They found that prolongation of the QT interval in first degree relatives of patients who suffered from DILQTS is greater than in first degree relatives of patients using an anti-arrhythmic drug for a long time without any sign of significant QT prolongation. A number of studies have since been carried out to study genetic variation in DILQTS patients, initially targeting candidate genes which cause cLQTS.

Genetic variation increases the risk of drug-induced arrhythmia

More than 700 mutations in at least 13 genes have been found to be associated with the cause of cLQTS. Six of these genes encode pore-forming ion channels (KCNQ1, KCNH2, SCN5A, KCNJ2, CACNA1C, and KCNJ5) and seven of these genes encode for ion channel subunits or regulatory proteins (KCNE1, KCNE2, ANKB, CAV3, SCN4B, AKAP9, and SNTA1). KCNQ1 and KCNE1 co-assemble to form IKs while KCNH2 and KCNE2 co-assemble to form IKr. A summary of the studies and genes investigated for association with drug-induced arrhythmias can be found in Table 3.

Table 3.

The genes and associated coding variants that have been found to be associated with drug-induced arrhythmias

Genes Associated phenotype Variants Functional assessment Studies
SCN5A LQT3 S1102Y Yes Splawski et al., 2002 23
G1844A, C1852T, T3748C Yes Yang et al., 2002 25
KCNH2 LQT2 C2350T Yes Yang et al., 2002 25
P347S Yes Paulussen et al., 2004 26
R1033W No Ramirez et al., 2012 30
KCNQ1 LQT1 C1747T Yes Yang et al., 2002 25
Y315C Yes Napolitano et al., 2000 24
KCNE1 LQT5 D85N Yes Paulussen et al., 2004 26 and Kaab et al., 2011 4
KCNE2 LQT6 T8A Yes Paulussen et al., 2004 26, Sesti et al., 2000 21, Abbott et al., 1999 22
NOS1AP QT interval rs10919035 Yes Jamshidi et al., 2012 42
CACNA1C LQT8 A1733V No Ramirez et al., 2012 30
AKAP9 LQT11 Q3531E No Ramirez et al., 2012 30
SNTA1 LQT12 T147N No Ramirez et al., 2012 30
KCND3 Brugada syndrome R566C No Ramirez et al., 2012 30
GPD1L Brugada syndrome V249M No Ramirez et al., 2012 30
RYR2 Catecholaminergic polymorphic ventricular tachycardia L555V, L2607P, E4361Q No Ramirez et al., 2012 30
CACNB2 Short QT syndrome M1V, I170V No Ramirez et al., 2012 30
KCNN3 Atrial fibrillation F315L No Ramirez et al., 2012 30
PPP2R3A F1000L No Ramirez et al., 2012 30
AKAP7 Q112R No Ramirez et al., 2012 30
APLP2 R504L No Ramirez et al., 2012 30
ATP2A2 S184C No Ramirez et al., 2012 30
AKAP6 V839A No Ramirez et al., 2012 30
ZFHX3 K3689E, T3640M, H3611Y, L741F, G117S No Ramirez et al., 2012 30
JPH3 R656W No Ramirez et al., 2012 30
CALR D165G No Ramirez et al., 2012 30
JPH2 V345L, T286A No Ramirez et al., 2012 30

LQTX, subtype of the congenital long QT syndrome;

Non-coding variant associated with dLQTS.

Sesti et al. screened the KCNE2 gene in 98 cases of drug-induced arrhythmia and 200 control subjects 21. They identified three sporadic mutations (M54T, I57T and A116V) and one more common rare variant (T8A). T8A was found in only 16 of 1010 (1.6%) control individuals and 1 of 230 (0.4%) index patients with LQTS. Functional and in silico analyses demonstrated that although the mutations produced reductions in current density, none of the three sporadic mutations altered the susceptibility to drug inhibition. On the other hand, only a 15% decrease in current density was observed with the T8A variant, and patients with this genotype have a normal QT interval at baseline 22. Interestingly, on drug exposure this variant increased the inhibitory effects of antibiotic treatment and demonstrated that common sequence variations can be clinically silent before drug exposure yet increase the risk of DILQTS.

A non-synonymous variant, S1102Y in the SCN5A gene, was associated with an increased risk of sudden cardiac death (SCD) in African-American adults and children although it is rarer or even absent in other ethnic groups. Splawski et al. screened cases of cardiac arrhythmia (predominantly due to medication) and S1102Y heterozygosity was associated with an eight-fold higher risk of arrhythmia than in non-carriers of the variant 23. Functional and in silico studies of Y1102 and computational analysis of simulated action potentials generated by S1102/Y1102 channels demonstrated prolonged repolarization and early after depolarizations (EADs) associated with the Y1102 variant.

At the same time, an early candidate gene study was performed in 92 subjects who had experienced drug-induced arrhythmia and 157 healthy subjects in three genes firmly associated with cLQTS. The study identified mutations in five of the 92 subjects 25; three of them were in the SCN5A gene (G1844A, C1852T, T3748C), one in the KCNQ1 gene (C1747T) 24 and one in the KCNH2 gene (C2350T). Four polymorphisms were also identified but there was no difference in the frequency between the cases and the controls 25.

A candidate gene study by Paulussen et al. screened the five main cLQTS associated genes in 32 drug-induced arrhythmia patients with confirmed TdP and identified three missense variants: T8A in KCNE2, D85N in KCNE1 and P347S in KCNH2 26. T8A and D85N have been implicated with drug-induced arrhythmias in earlier studies as well as in patients suffering from QT prolongation 21,22. Three additional non-synonymous variations were found in both the controls and the cases with similar allele frequencies in the two populations in the KCNE1, KCNH2 and SCN5A genes. These variants have also been previously reported in earlier studies in patients suffering from cLQTS 25,2729.

Kaab et al. confirmed the association of the KCNE1-D85N variant and drug-induced arrhythmia in 176 cases and 207 drug-exposed controls (individuals without QT prolongation when exposed to culpable medications) and 837 controls from the general population 4. The study included 1424 single nucleotide polymorphisms (SNPs) in 18 candidate genes (which included ion channels and other high priority candidate genes) for which the subjects were genotyped. The most statistically significant associated SNP was rs7295250 in the CACNA1C gene (odds ratio 1.88, P = 7.62 × 10−4). The strongest effect size within the study was detected for D85N (odds ratio 8.88, P = 1.95 × 10−5). The follow-up validation study failed to replicate the initial association although a trend for the D58N variant was observed (P = 0.58).

A more recent study by Ramirez et al. utilized a next generation sequencing panel of 79 genes associated with arrhythmia syndromes including 13 genes which have been previously associated with cLQTS, nine additional genes shown to be involved in congenital short QT syndrome (cSQTS) and Brugada syndrome and nine other genes related to familial arrhythmia syndromes 30. In 11/31 DILQTS (36%) subjects a novel missense mutation in genes known to be associated with congenital arrhythmia was found. In 6/26 (23%) DILQTS Caucasian subjects a conserved deleterious variation or an already identified congenital arrhythmia mutation was found. These variants were found in less than 2% of the individuals sequenced as part of the 1000 Genomes project. The conclusion from this study was that the rare variants in arrhythmia related genes, not just cLQTS genes, are responsible for a significant proportion of drug-induced arrhythmia cases. These conclusions may be limited, however, by the different platforms and depth of sequencing in the case and control groups. Overall, however, around 10% of cases may be associated with rare variation in the cLQTS genes 5.

Common variation in genes associated with QT prolongation has also been investigated as a potential cause of drug-induced arrhythmias. The NOS1AP (CAPON) gene has been repeatedly associated with QT prolongation in healthy individuals 3133 and it has also been associated with severity in patients suffering from cLQTS 34. NOS1AP is a regulator of neuronal nitric oxide synthase (nNOS), which in turn regulates the intracellular levels of calcium and myocyte contraction in the heart 3537. NOS1AP is thought to alter cardiac repolarization by interaction with nNOS by blocking L-type calcium channels 3841. This may explain the association of NOS1AP gene variants with prolongation of the QT interval and TdP.

Jamshidi et al. demonstrated that a common variant (rs10919035, odds ratio 5.5, P = 3.0 × 104) in the NOS1AP gene which has been previously associated with the QT interval prolongation plays an important role in the risk of amiodarone-induced drug-induced arrhythmia 42. The study included 86 cases, 192 controls and 68 drug-exposed individuals with no QT prolongation. The frequency of the common variant in cases was 27.8% and in controls 7.1%. This supports the theory of repolarization reserve and its role in DITDP.

Drug-induced arrhythmia associated with drug metabolism

Anti-arrhythmic drugs that are known to cause drug-induced arrhythmia as an adverse reaction include mainly class III anti-arrhythmic agents which prolong the QT interval by blocking the IKr current. Variability in the response to drug therapy can be observed in patients. This variability may result not only from end-organ effects but also from drug concentration differences 43. Prolonged QT intervals following drug therapy in some individuals can therefore potentially be caused by common variants that alter drug metabolism thereby increasing drug concentration 44,45.

There are many genetic variants responsible for altered biological activity of enzymes belonging to the hepatic cytochrome P450 enzymes such as CYP3A4, CYP3A5 and CYP3A7. CYP2D6 activity is absent in 5–10% of Caucasian and African individuals due to underlying genetic variants in the CYP2D6 gene. This enzyme is responsible for metabolizing thioridazine, an anti-psychotic drug that has been discontinued in the UK due to its risk of TdP. It is therefore possible that the dysfunctional metabolic pathway may lead to QT prolongation and perhaps drug-induced arrhythmia 46. Paulussen et al. reported an individual with already compromised repolarization reserve due to a KCNH2-P347S mutation 26. This subject received a combination of drugs that both prolonged the QT interval 47: cisapride and clarithromycin. In addition, both of these drugs are metabolized by the same enzyme which is inhibited by clarithromycin thereby slowing the metabolism of cisapride. This exacerbates IKr blockade by increasing the concentrations of circulating cisapride in plasma 48 and offers a model for how genetic variability in the capacity for drug metabolism may work. As yet, however, there are no genetic data to support metabolic predisposition as a cause for TdP.

Genome-wide association studies (GWAS) and drug-induced arrhythmia

More recently the GWAS approach has been employed to investigate the association of the genetic risk factors across the genome with disease. GWASs that focus on common disorders have the advantage of including thousands of cases, whereas GWASs that focus on rarer disorders such as drug-induced adverse reactions do not involve many cases. Nonetheless, there are phenotypes where the GWAS approach has been successful with less than one hundred individuals 49,50.

A recent meta-analysis of 10 cohorts including 33 781 participants of European descent was performed to examine whether common genetic variants modify the effects of exposure to drugs on QT interval. The study examined four drug classes previously associated with QT prolongation, with the estimated prevalence of drug exposure highest for diuretics (13.6%). The study by Avery et al. found no genome-wide significant interactions for any of the four drug classes 51. An analysis limited to common variants identified in previously reported GWAS of QT interval yielded similarly null results, as did one restricted to recent pharmacogenomic studies of QT and drug-induced QT prolongation. Statistical power simulations suggested that at least for the diuretics class, if the genetic variation was common, the study had 80% power to detect an effect of 3.25 ms. However, this size of genetic effect is typically outside the range usually observed for QT interval. Furthermore, the study did not address the potential for bias related to duration of drug use, particularly as participants taking the drugs for years or decades are those least likely to have experienced side effects. Nevertheless, the results do suggest that increasing the number of measures per participant in longitudinal modelling may help to increase statistical power.

A more recent effort to investigate the role of common genetic variation on risk of DITdP in a GWAS concluded that common genetic variation with a strong effect-size did not play a role in the risk of drug-induced arrhythmias across all drug groups 6. The study included 264 Caucasian cases selected from two groups, the Trans-Atlantic Alliance against Sudden Death (the Fondation Leducq) and the Drug-induced Arrhythmia Risk Evaluation (DARE) study and 5350 controls, some of whom received medications showing no considerable QT interval prolongation and others general population controls. There were no genome-wide associations between common SNPs or genes and the risk of DITdP.

Conclusions

DILQTS remains a significant cause of morbidity and mortality, and is the major reason for the withdrawal of drugs from the market. Knowledge of rare and common variants that may modulate patients' susceptibility to adverse drug reactions such as DILQTS may allow clinicians to identify prospectively subpopulations at risk in the future. A number of genetic studies have determined that DILQTS is predisposed to by ethnically specific common variants found in genes modulating the length of the QT interval (SCN5A S1102Y, KCNE1 D85N and NOS1AP rs10919035). Rare variation in arrhythmia genes also appears to have a role in approximately 10% of cases. Attempts to investigate the role of common variants in a genome wide analysis has not been successful and will most likely require recruitment of substantially larger number of cases, with drug-specific analyses 6. However, the use of whole exome sequencing in subjects with DILQTS might be worth considering for future studies as it would reveal novel and low frequency alleles that may carry a genetic risk for a disease, whereas GWAS can only interrogate risk markers that are more common in the population. Recent breakthroughs in stem cell biology have led to a number of papers showing the possibility of using patient-derived induced pluripotent stem cells to further our understanding of the underlying mechanisms of pro-arrhythmia 5254. These studies may identify novel target genes involved in drug sensitivity and pro-arrhythmia which may warrant investigation for underlying genetic variants that can modulate their expression.

Competing Interests

Research funds were received from the international Serious Adverse Events Consortium.

YJ is supported by the British Heart Foundation (Project Grant 29615).

References

  • 1.Roden DM. Long QT syndrome: reduced repolarization reserve and the genetic link. J Intern Med. 2006;259:59–69. doi: 10.1111/j.1365-2796.2005.01589.x. [DOI] [PubMed] [Google Scholar]
  • 2.Kozik TM, Wung SF. Cardiac arrest from acquired long QT syndrome: a case report. Heart Lung. 2009;38:238–242. doi: 10.1016/j.hrtlng.2008.08.002. [DOI] [PubMed] [Google Scholar]
  • 3.Saenen JB, Vrints CJ. Molecular aspects of the congenital and acquired long QT syndrome: clinical implications. J Mol Cell Cardiol. 2008;44:633–646. doi: 10.1016/j.yjmcc.2008.01.006. [DOI] [PubMed] [Google Scholar]
  • 4.Kaab S, Crawford DC, Sinner MF, Behr ER, Kannankeril PJ, Wilde AA, Bezzina CR, Schulze-Bahr E, Guicheney P, Bishopric NH, Myerburg RJ, Schott JJ, Pfeufer A, Beckmann BM, Martens E, Zhang T, Stallmeyer B, Zumhagen S, Denjoy I, Bardai A, Van Gelder IC, Jamshidi Y, Dalageorgou C, Marshall V, Jeffery S, Shakir S, Camm AJ, Steinbeck G, Perz S, Lichtner P, Meitinger T, Peters A, Wichmann HE, Ingram C, Bradford Y, Carter S, Norris K, Ritchie MD, George AL, Jr, Roden DM. A large candidate gene survey identifies the KCNE1 D85N polymorphism as a possible modulator of drug-induced torsades de pointes. Circ Cardiovasc Genet. 2011;5:91–99. doi: 10.1161/CIRCGENETICS.111.960930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Behr ER, Roden D. Drug-induced arrhythmia: pharmacogenomic prescribing? Eur Heart J. 2013;34:89–95. doi: 10.1093/eurheartj/ehs351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Behr ER, January C, Schulze-Bahr E, Grace AA, Kaab S, Fiszman M, Gathers S, Buckman S, Youssef A, Pirmohamed M, Roden D. The International Serious Adverse Events Consortium (iSAEC) phenotype standardization project for drug-induced torsades de pointes. Eur Heart J. 2013;34:1958–1963. doi: 10.1093/eurheartj/ehs172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dekker JM, Crow RS, Hannan PJ, Schouten EG, Folsom AR ARIC Study. Heart rate-corrected QT interval prolongation predicts risk of coronary heart disease in black and white middle-aged men and women: the ARIC study. J Am Coll Cardiol. 2004;43:565–571. doi: 10.1016/j.jacc.2003.09.040. [DOI] [PubMed] [Google Scholar]
  • 8.Nakanishi S, Yamada M, Hattori N, Suzuki G. Relation between QT duration and mortality in an elderly Japanese population. Am J Cardiol. 2004;93:1182–1185. doi: 10.1016/j.amjcard.2004.01.055. [DOI] [PubMed] [Google Scholar]
  • 9.Anttonen O, Junttila MJ, Rissanen H, Reunanen A, Viitasalo M, Huikuri HV. Prevalence and prognostic significance of short QT interval in a middle-aged Finnish population. Circulation. 2007;116:714–720. doi: 10.1161/CIRCULATIONAHA.106.676551. [DOI] [PubMed] [Google Scholar]
  • 10.Zhang Y, Post WS, Blasco-Colmenares E, Dalal D, Tomaselli GF, Guallar E. Electrocardiographic QT interval and mortality: a meta-analysis. Epidemiology. 2011;22:660–670. doi: 10.1097/EDE.0b013e318225768b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jons C, O-Uchi J, Moss AJ, Reumann M, Rice JJ, Goldenberg I, Zareba W, Wilde AA, Shimizu W, Kanters JK, McNitt S, Hofman N, Robinson JL, Lopes CM. Use of mutant-specific ion channel characteristics for risk stratification of long QT syndrome patients. Sci Transl Med. 2011;3:76ra28. doi: 10.1126/scitranslmed.3001551. [DOI] [PubMed] [Google Scholar]
  • 12.Newton-Cheh C, Shah R. Genetic determinants of QT interval variation and sudden cardiac death. Curr Opin Genet Dev. 2007;17:213–221. doi: 10.1016/j.gde.2007.04.010. [DOI] [PubMed] [Google Scholar]
  • 13.Roden DM. Cardiovascular pharmacogenomics: the future of cardiovascular therapeutics? Can J Cardiol. 2013;29:58–66. doi: 10.1016/j.cjca.2012.07.845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Noble D, Tsien RW. Outward membrane currents activated in the plateau range of potentials in cardiac Purkinje fibres. J Physiol. 1969;200:205–231. doi: 10.1113/jphysiol.1969.sp008689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sanguinetti MC, Jurkiewicz NK. Two components of cardiac delayed rectifier K+ current. Differential sensitivity to block by class III antiarrhythmic agents. J Gen Physiol. 1990;96:195–215. doi: 10.1085/jgp.96.1.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Balser JR, Bennett PB, Roden DM. Time-dependent outward current in guinea pig ventricular myocytes. Gating kinetics of the delayed rectifier. J Gen Physiol. 1990;96:835–863. doi: 10.1085/jgp.96.4.835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fenichel RR, Malik M, Antzelevitch C, Sanguinetti M, Roden DM, Priori SG, Ruskin JN, Lipicky RJ, LR Cantilena Independent Academic Task Force. Drug-induced torsades de pointes and implications for drug development. J Cardiovasc Electrophysiol. 2004;15:475–495. doi: 10.1046/j.1540-8167.2004.03534.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Roden DM. Taking the ‘idio’ out of ‘idiosyncratic’: predicting torsades de pointes. Pacing Clin Electrophysiol. 1998;21:1029–1034. doi: 10.1111/j.1540-8159.1998.tb00148.x. [DOI] [PubMed] [Google Scholar]
  • 19.Xiao L, Xiao J, Luo X, Lin H, Wang Z, Nattel S. Feedback remodeling of cardiac potassium current expression: a novel potential mechanism for control of repolarization reserve. Circulation. 2008;118:983–992. doi: 10.1161/CIRCULATIONAHA.107.758672. [DOI] [PubMed] [Google Scholar]
  • 20.Kannankeril PJ, Roden DM, Norris KJ, Whalen SP, George AL, Jr, Murray KT. Genetic susceptibility to acquired long QT syndrome: pharmacologic challenge in first-degree relatives. Heart Rhythm. 2005;2:134–140. doi: 10.1016/j.hrthm.2004.10.039. [DOI] [PubMed] [Google Scholar]
  • 21.Sesti F, Abbott GW, Wei J, Murray KT, Saksena S, Schwartz PJ, Priori SG, Roden DM, George AL, Jr, Goldstein SA. A common polymorphism associated with antibiotic-induced cardiac arrhythmia. Proc Natl Acad Sci U S A. 2000;97:10613–10618. doi: 10.1073/pnas.180223197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Abbott GW, Sesti F, Splawski I, Buck ME, Lehmann MH, Timothy KW, Keating MT, Goldstein SA. MiRP1 forms IKr potassium channels with HERG and is associated with cardiac arrhythmia. Cell. 1999;97:175–187. doi: 10.1016/s0092-8674(00)80728-x. [DOI] [PubMed] [Google Scholar]
  • 23.Splawski I, Timothy KW, Tateyama M, Clancy CE, Malhotra A, Beggs AH, Cappuccio FP, Sagnella GA, Kass RS, Keating MT. Variant of SCN5A sodium channel implicated in risk of cardiac arrhythmia. Science. 2002;297:1333–1336. doi: 10.1126/science.1073569. [DOI] [PubMed] [Google Scholar]
  • 24.Napolitano C, Schwartz PJ, Brown AM, Ronchetti E, Bianchi L, Pinnavaia A, Acquaro G, Priori SG. Evidence for a cardiac ion channel mutation underlying drug-induced QT prolongation and life-threatening arrhythmias. Am J Physiol Heart Circ Physiol. 2000;279:H300–311. doi: 10.1111/j.1540-8167.2000.tb00033.x. [DOI] [PubMed] [Google Scholar]
  • 25.Yang P, Kanki H, Drolet B, Yang T, Wei J, Viswanathan PC, Hohnloser SH, Shimizu W, Schwartz PJ, Stanton M, Murray KT, Norris K, George AL, Jr, Roden DM. Allelic variants in long-QT disease genes in patients with drug-associated torsades de pointes. Circulation. 2002;105:1943–1948. doi: 10.1161/01.cir.0000014448.19052.4c. [DOI] [PubMed] [Google Scholar]
  • 26.Paulussen AD, Gilissen RA, Armstrong M, Doevendans PA, Verhasselt P, Smeets HJ, Schulze-Bahr E, Haverkamp W, Breithardt G, Cohen N, Aerssens J. Genetic variations of KCNQ1, KCNH2, SCN5A, KCNE1, and KCNE2 in drug-induced long QT syndrome patients. J Mol Med. 2004;82:182–188. doi: 10.1007/s00109-003-0522-z. [DOI] [PubMed] [Google Scholar]
  • 27.Lai LP, Deng CL, Moss AJ, Kass RS, Liang CS. Polymorphism of the gene encoding a human minimal potassium ion channel (minK) Gene. 1994;151:339–340. doi: 10.1016/0378-1119(94)90685-8. [DOI] [PubMed] [Google Scholar]
  • 28.Tesson F, Donger C, Denjoy I, Berthet M, Bennaceur M, Petit C, Coumel P, Schwarts K, Guicheney P. Exclusion of KCNE1 (IsK) as a candidate gene for Jervell and Lange-Nielsen syndrome. J Mol Cell Cardiol. 1996;28:2051–2055. doi: 10.1006/jmcc.1996.0198. [DOI] [PubMed] [Google Scholar]
  • 29.Iwasa H, Itoh T, Nagai R, Nakamura Y, Tanaka T. Twenty single nucleotide polymorphisms (SNPs) and their allelic frequencies in four genes that are responsible for familial long QT syndrome in the Japanese population. J Hum Genet. 2000;45:182–183. doi: 10.1007/s100380050207. [DOI] [PubMed] [Google Scholar]
  • 30.Ramirez AH, Shaffer CM, Delaney JT, Sexton DP, Levy SE, Rieder MJ, Nickerson DA, George AL, Jr, Roden DM. Novel rare variants in congenital cardiac arrhythmia genes are frequent in drug-induced torsades de pointes. Pharmacogenomics J. 2012;13:325–329. doi: 10.1038/tpj.2012.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Newton-Cheh C, Eijgelsheim M, Rice KM, de Bakker PI, Yin X, Estrada K, Bis JC, Marciante K, Rivadeneira F, Noseworthy PA, Sotoodehnia N, Smith NL, Rotter JI, Kors JA, Witteman JC, Hofman A, Heckbert SR, O'Donnell CJ, Uitterlinden AG, Psaty BM, Lumley T, Larson MG, Stricker BH. Common variants at ten loci influence QT interval duration in the QTGEN Study. Nat Genet. 2009;41:399–406. doi: 10.1038/ng.364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pfeufer A, Sanna S, Arking DE, Muller M, Gateva V, Fuchsberger C, Ehret GB, Orrú M, Pattaro C, Köttgen A, Perz S, Usala G, Barbalic M, Li M, Pütz B, Scuteri A, Prineas RJ, Sinner MF, Gieger C, Najjar SS, Kao WH, Mühleisen TW, Dei M, Happle C, Möhlenkamp S, Crisponi L, Erbel R, Jöckel KH, Naitza S, Steinbeck G, Marroni F, Hicks AA, Lakatta E, Müller-Myhsok B, Pramstaller PP, Wichmann HE, Schlessinger D, Boerwinkle E, Meitinger T, Uda M, Coresh J, Kääb S, Abecasis GR, Chakravarti A. Common variants at ten loci modulate the QT interval duration in the QTSCD Study. Nat Genet. 2009;41:407–414. doi: 10.1038/ng.362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nolte IM, Wallace C, Newhouse SJ, Waggott D, Fu J, Soranzo N, Gwilliam R, Deloukas P, Savelieva I, Zheng D, Dalageorgou C, Farrall M, Samani NJ, Connell J, Brown M, Dominiczak A, Lathrop M, Zeggini E, LV Wain, Wellcome Trust Case Control Consortium. DCCT/EDIC Research Group. Newton-Cheh C, Eijgelsheim M, Rice K, de Bakker PI, QTGEN consortium. Pfeufer A, Sanna S, Arking DE, QTSCD consortium. Asselbergs FW, Spector TD, Carter ND, Jeffery S, Tobin M, Caulfield M, Snieder H, Paterson AD, Munroe PB, Jamshidi Y. Common genetic variation near the phospholamban gene is associated with cardiac repolarisation: meta-analysis of three genome-wide association studies. PLoS ONE. 2009;4:e6138. doi: 10.1371/journal.pone.0006138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tomas M, Napolitano C, De Giuli L, Bloise R, Subirana I, Malovini A, Bellazzi R, Arking DE, Marban E, Chakravarti A, Spooner PM, Priori SG. Polymorphisms in the NOS1AP gene modulate QT interval duration and risk of arrhythmias in the long QT syndrome. J Am Coll Cardiol. 2010;55:2745–2752. doi: 10.1016/j.jacc.2009.12.065. [DOI] [PubMed] [Google Scholar]
  • 35.Barouch LA, Harrison RW, Skaf MW, Rosas GO, Cappola TP, Kobeissi ZA, Hobai IA, Lemmon CA, Burnett AL, O′Rourke B, Rodriguez ER, Huang PL, Lima JA, Berkowitz DE, Hare JM. Nitric oxide regulates the heart by spatial confinement of nitric oxide synthase isoforms. Nature. 2002;416:337–339. doi: 10.1038/416337a. [DOI] [PubMed] [Google Scholar]
  • 36.Burkard N, Rokita AG, Kaufmann SG, Hallhuber M, Wu R, Hu K, Hofmann U, Bonz A, Frantz S, Cartwright EJ, Neyses L, Maier LS, Maier SK, Renné T, Schuh K, Ritter O. Conditional neuronal nitric oxide synthase overexpression impairs myocardial contractility. Circ Res. 2007;100:e32–44. doi: 10.1161/01.RES.0000259042.04576.6a. [DOI] [PubMed] [Google Scholar]
  • 37.Oceandy D, Cartwright EJ, Emerson M, Prehar S, Baudoin FM, Zi M, Alatwi N, Venetucci L, Schuh K, Williams JC, Armesilla AL, Neyses L. Neuronal nitric oxide synthase signaling in the heart is regulated by the sarcolemmal calcium pump 4b. Circulation. 2007;115:483–492. doi: 10.1161/CIRCULATIONAHA.106.643791. [DOI] [PubMed] [Google Scholar]
  • 38.Chang KC, Barth AS, Sasano T, Kizana E, Kashiwakura Y, Zhang Y, Foster DB, Marbán E. CAPON modulates cardiac repolarization via neuronal nitric oxide synthase signaling in the heart. Proc Natl Acad Sci U S A. 2008;105:4477–4482. doi: 10.1073/pnas.0709118105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ueda K, Valdivia C, Medeiros-Domingo A, Tester DJ, Vatta M, Farrugia G, Ackerman MJ, Makielski JC. Syntrophin mutation associated with long QT syndrome through activation of the nNOS-SCN5A macromolecular complex. Proc Natl Acad Sci U S A. 2008;105:9355–9360. doi: 10.1073/pnas.0801294105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R, Napolitano C, Schwartz PJ, Joseph RM, Condouris K, Tager-Flusberg H, Priori SG, Sanguinetti MC, Keating MT. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell. 2004;119:19–31. doi: 10.1016/j.cell.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 41.Splawski I, Timothy KW, Decher N, Kumar P, Sachse FB, Beggs AH, Sanguinetti MC, Keating MT. Severe arrhythmia disorder caused by cardiac L-type calcium channel mutations. Proc Natl Acad Sci U S A. 2005;102:8089–8096. doi: 10.1073/pnas.0502506102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jamshidi Y, Nolte IM, Dalageorgou C, Zheng D, Johnson T, Bastiaenen R, Ruddy S, Talbott D, Norris KJ, Snieder H, George AL, Marshall V, Shakir S, Kannankeril PJ, Munroe PB, Camm AJ, Jeffery S, Roden DM, Behr ER. Common variation in the NOS1AP gene is associated with drug-induced QT prolongation and ventricular arrhythmia. J Am Coll Cardiol. 2012;60:841–850. doi: 10.1016/j.jacc.2012.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Roden DM, Johnson JA, Kimmel SE, Krauss RM, Medina MW, Shuldiner A, Wilke RA. Cardiovascular pharmacogenomics. Circ Res. 2011;109:807–820. doi: 10.1161/CIRCRESAHA.110.230995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kannankeril PJ, Roden DM. Drug-induced long QT and torsade de pointes: recent advances. Curr Opin Cardiol. 2007;22:39–43. doi: 10.1097/HCO.0b013e32801129eb. [DOI] [PubMed] [Google Scholar]
  • 45.Johnson JA, Cavallari LH, Beitelshees AL, Lewis JP, Shuldiner AR, Roden DM. Pharmacogenomics: application to the management of cardiovascular disease. Clin Pharmacol Ther. 2011;90:519–531. doi: 10.1038/clpt.2011.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.LLerena A, Berecz R, de la Rubia A, Dorado P. QTc interval lengthening is related to CYP2D6 hydroxylation capacity and plasma concentration of thioridazine in patients. J Psychopharmacol. 2002;16:361–364. doi: 10.1177/026988110201600411. [DOI] [PubMed] [Google Scholar]
  • 47.Mohammad S, Zhou Z, Gong Q, January CT. Blockage of the HERG human cardiac K+ channel by the gastrointestinal prokinetic agent cisapride. Am J Physiol. 1997;273(5 Pt 2):H2534–2538. doi: 10.1152/ajpheart.1997.273.5.H2534. [DOI] [PubMed] [Google Scholar]
  • 48.Piquette RK. Torsade de pointes induced by cisapride/clarithromycin interaction. Ann Pharmacother. 1999;33:22–26. doi: 10.1345/aph.18107. [DOI] [PubMed] [Google Scholar]
  • 49.Daly AK, Donaldson PT, Bhatnagar P, Shen Y, Pe'er I, Floratos A, Daly MJ, Goldstein DB, John S, Nelson MR, Graham J, Park BK, Dillon JF, Bernal W, Cordell HJ, Pirmohamed M, Aithal GP, Day CP DILIGEN Study; International SAE Consortium. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet. 2009;41:816–819. doi: 10.1038/ng.379. [DOI] [PubMed] [Google Scholar]
  • 50.McCormack M, Alfirevic A, Bourgeois S, Farrell JJ, Kasperaviciute D, Carrington M, Sills GJ, Marson T, Jia X, de Bakker PI, Chinthapalli K, Molokhia M, Johnson MR, O′Connor GD, Chaila E, Alhusaini S, Shianna KV, Radtke RA, Heinzen EL, Walley N, Pandolfo M, Pichler W, Park BK, Depondt C, Sisodiya SM, Goldstein DB, Deloukas P, Delanty N, Cavalleri GL, Pirmohamed M. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med. 2011;364:1134–1143. doi: 10.1056/NEJMoa1013297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Avery CL, Sitlani CM, Arking DE, Arnett DK, Bis JC, Boerwinkle E, Buckley BM, Ida Chen YD, de Craen AJ, Eijgelsheim M, Enquobahrie D, Evans DS, Ford I, Garcia ME, Gudnason V, Harris TB, Heckbert SR, Hochner H, Hofman A, Hsueh WC, Isaacs A, Jukema JW, Knekt P, Kors JA, Krijthe BP, Kristiansson K, Laaksonen M, Liu Y, Li X, Macfarlane PW, Newton-Cheh C, Nieminen MS, Oostra BA, Peloso GM, Porthan K, Rice K, Rivadeneira FF, Rotter JI, Salomaa V, Sattar N, Siscovick DS, Slagboom PE, Smith AV, Sotoodehnia N, Stott DJ, Stricker BH, Stürmer T, Trompet S, Uitterlinden AG, van Duijn C, Westendorp RG, Witteman JC, Whitsel EA, Psaty BM. Drug–gene interactions and the search for missing heritability: a cross-sectional pharmacogenomics study of the QT interval. Pharmacogenomics J. 2013 doi: 10.1038/tpj.2013.4. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Egashira T, Yuasa S, Suzuki T, Aizawa Y, Yamakawa H, Matsuhashi T, Ohno Y, Tohyama S, Okata S, Seki T, Kuroda Y, Yae K, Hashimoto H, Tanaka T, Hattori F, Sato T, Miyoshi S, Takatsuki S, Murata M, Kurokawa J, Furukawa T, Makita N, Aiba T, Shimizu W, Horie M, Kamiya K, Kodama I, Ogawa S, Fukuda K. Disease characterization using LQTS-specific induced pluripotent stem cells. Cardiovasc Res. 2012;95:419–429. doi: 10.1093/cvr/cvs206. [DOI] [PubMed] [Google Scholar]
  • 53.Lahti AL, Kujala VJ, Chapman H, Koivisto AP, Pekkanen-Mattila M, Kerkela E, Hyttinen J, Kontula K, Swan H, Conklin BR, Yamanaka S, Silvennoinen O, Aalto-Setälä K. Model for long QT syndrome type 2 using human iPS cells demonstrates arrhythmogenic characteristics in cell culture. Dis Model Mech. 2012;5:220–230. doi: 10.1242/dmm.008409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O, Winterstern A, Feldman O, Gepstein A, Arbel G, Hammerman H, Boulos M, Gepstein L. Modelling the long QT syndrome with induced pluripotent stem cells. Nature. 2011;471:225–229. doi: 10.1038/nature09747. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES