Submit manuscript...
Journal of
eISSN: 2473-0831

Analytical & Pharmaceutical Research

Review Article Volume 7 Issue 2

Cardiotoxicity of H1-antihistamines

Loipa Galán Martínez, Gabriel Emilio Curbelo González

Correspondence: Loipa Galán Martínez, Laboratorio de Electrofisiología, Departamento de Investigaciones, Instituto de Cardiología y Cirugía Cardiovascular, Paseo y 17, Vedado, CP 10400, La Habana, Cuba

Received: March 01, 2018 | Published: April 12, 2018

Citation: Martínez LG, González GEC. Cardiotoxicity of H1-antihistamines. J Anal Pharm Res. 2018;7(2):197-201. DOI: 10.15406/japlr.2018.07.00226

Download PDF

Abstract

The H1-antihistamines are drugs that have been developed and marketed to treat noncardiovascular diseases and these drugs have been known to induce a number of untoward cardiovascular effects. This review summarizes current literature to evaluate the cardiotoxicity of H1-antihistamines.

Keywords: arrhythmia, cardiotoxicity, cardiovascular, H1-antihistamines, QT prolongation

Abbreviations

H1A, H1-antihistamines; hERG, human ether-a-go-go–related gene

Introduction

Today the H-1antihistamines (H1A) are among the most widely used drugs worldwide to treat various allergic diseases, for which its effectiveness has been proven, such as allergic rhinitis, conjunctivitis and urticaria.1 Also H1A are used for other purposes such as prevention/treatment of disorders related to activation of the vomiting centre and sedation.2 H1A act as inverse agonists that combine and stabilize with the inactive conformation of the H1 receptor, shifting the equilibrium to the inactive state. Through this mechanism of action, these compounds can regulate allergic inflammation.3

While sedation and antimuscarinic effects are the most frequent adverse drug reactions for H1A, cardiac toxicity represents a very rare but severe complication.1–6 First generation sedative antihistamines-e.g., diphenhydramine, which causes marked sedation, central nervous system dysfunction, and anticholinergic adverse effects- are still widely used in several countries,6,7 where they are a major cause of drug toxicity. Second generation H1A were introduced in the 1980s as non-sedative and specific H1A.1,3,4,6 However, after approximately 10 years of worldwide use, H1A such as astemizole and terfenadine, were found to cause prolonged QT interval in the surface electrocardiogram and were withdrawn from the market due to the risk of sudden cardiac death and ventricular tachyarrhythmia, associated with QT prolongation.1,4,6 It has prompted a reexamination of the possible cardiac adverse effects exerted by older compounds belonging to this therapeutic class of drugs.6

Clinical evidence

There is a good amount of non-cardiovascular drugs that produce undesirable side effects in cardiovascular physiology, such as antibiotics8–10 and H1A.1,2,4–6,10 Many AH1 are available worldwide today (Table 1), and new types are developed nowadays.1,3,11 Several clinically useful drugs are able to antagonize the effects of histamine at the level of H1 receptors were developed in the fourth and fifth decades of the last century.1,3,12 These included chlorpheniramine, brompheniramine, hydroxyzine, and diphenhydramine. To improve selectivity for H1 histamine receptors and to minimize the well-known sedative effects of these “first generation” molecules, the so-called “second generation” H1A were introduced in the mid 1980s. Terfenadine was the first of these to be marketed, astemizole was the second of the newer nonsedating drugs to be marketed, and it has been widely prescribed, although less than terfenadine. Also loratadine and cetirizine have been available for several years; these drugs are also less sedating and have been approved for marketing,1,3,4,6,11,12 these two nonsedating H1A at doses three to six times higher than clinically recommended for treatment of allergic conditions failed to prolong QT intervals in normal subjects.4,6,11,12 The second generation H1A remaining in use are free from potential cardiac adverse effects.1 Third generation H1A are the active enantiomer (levocetirizine) or metabolite (desloratadine and fexofenadine) derivatives of second generation drugs. These drugs were intended to be more efficacies with fewer adverse drug reactions. However, there is little evidence for any advantage of levocetirizine or desloratadine, compared to cetirizine or loratadine, respectively.5 The terms third generation, new generation, or next generation have been used to market some new H1A; however, this designation should be reserved for clinically advantageous H1A designed with the use of molecular techniques that might be available in the future.13

Generic name of H1A (Brand name)

Chemical class

Cardiotoxicity

Refs

First-generation

Doxepin (SINEQUAN)

Tricyclic Dibenzoxepin

Yes

20–22

Carbinoxamine maleate (RONDEC, others)

Ethanolamines

?

No reported

Clemastine fumarate (TAVIST, others)

Yes

23,24

Diphenhydramine (BENADRYL, NYTOL, UNISOM, SOMINEX, DIMEDROL, DAEDALON)

Yes (dose-dependent)

14,15,25–30

Dimenhydrinate (DRAMAMINE, others)

Yes

65,31–33

Pyrilamine maleate (POLY–HISTINE-D)

Ethylenediamines

Yes (overdose)

32,33

Tripelennamine (PBZ)

?

No reported

Chlorpheniramine maleate (CHLOR-TRIMETON, others)

Alkylamines

Yes (overdose)

23,33,35–38

Brompheniramine maleate (BROMPHEN, others)

Yes (non clinical)

39

Hydroxyzine (ATARAX, VISTARIL, ATERAX, ALAMON, DURRAX, EQUIPOSE, MASMORAN, ORGATRAX, PAXISTIL QUIES, TRAN-Q, TRANQUIZINE)

Piperazines

Yes

16–18,40–45

Cyclizine (MAREZINE)

?

No reported

Meclizine (ANTIVERT, others)

?

No reported

Promethazine (PHENERGAN, others)

Phenothiazine

Yes

46,47

Cyproheptadine (PERIACTIN)

Piperidines

No

42

Phenindamine tartrate (NOLAHIST)

?

No reported

Second-generation

Acrivastine (SEMPREX-D)

Alkylamine

?

No reported

Cetirizine (ZYRTEC)

Piperazine

No

2,48–50

Azelastine (ASTELIN)

Phthalazinone

Yes

52,53

Levocabastine (LIVOSTIN)

Piperidines

No

53

Loratadine (CLARITIN)

No

2,54,55

Desloratadine (CLARINEX, AERIUS)

No

56

Ebastine (EBASTEL)

Yes

2

Mizolastine (MIZOLLEN)

No

57–59

Fexofenadine (ALLEGRA, TELFAST)

Yes (one case reported)

60,61

Table 1 Cardiotoxicity associated with the consumption of first- and second-generation antihistamines

In 1986, reports began to appear indicating that patients who took terfenadine or astemizole could develop a classical form of ventricular arrhythmia, Torsades de pointes. This caused that these drugs were withdrawn from the market.4,6,11,12 Older first generation H1A are widely sold in most countries as “over the counter” or prescribed medications and are frequently implicated in accidental or intentional poisoning. The issue of the potential cardiotoxicity of older antihistamines has been raised by recent studies. Some first generation H1A potentially cause dose-related cardiac adverse effects, including sinus tachycardia, reflex tachycardia, supraventricular arrhythmias, and after intentional large overdose, for example, diphenhydramine 0.5 to 1 g, prolongation of the QT interval with ventricular arrhythmias and Torsades de pointes has been documented.14 Therapeutic doses of diphenhydramine were also effective in lengthening cardiac repolarization in vivo in normal volunteers and in patients undergoing angioplasty.15 Arrhythmogenic properties in vivo have also been described for promethazine, pheniramine and chlorpheniramine (Table 1).6

Recently, Poluzzi et al.2 reported increased risk of ventricular tachyarrhythmia for current use of cyclizine, dimetindene and ebastine; while cetirizine, fexofenadine and loratadine were in absence of high risk of ventricular tachyarrhythmia.2 Hydroxyzine, a sedative agent, which shares with cyclizine the molecular core (diphenyl-methyl piperazine), many receptor activities and some indications, recently received restrictions of use (i.e. limitations in daily dose and duration of exposure) by the European Medicine Agency for the proarrhythmic risk16 following pharmacovigilance findings,17 which confirmed evidence of potassium channel blocking properties.18 Serious cardiotoxic events occur very rarely during assumption of H1A; in a UK study involving approximately 200 000 people who received 500 000 prescriptions of five nonsedating H1A: acrivastine, astemizole, cetirizine, loratadine and terfenadine, it has been estimated that the absolute effect is quite low, requiring 57,000 prescriptions, or 5300 person-years of use, for one case to occur.19 Table 120–61 lists the H1A drugs that are available and summarizes the reported cardiotoxicity for some of them.

Molecular target for H1A-induced cardiotoxicity

Cardiac toxicity of H1A does not occur through the H1 receptor, and is not a class effect.1,3,4,6,11,12 Rather, it is due to blockage of cardiac ion currents, such as potassium and sodium channel.6,12 The most important molecular mechanism of QT prolonging drugs has been shown to be the blockade of the delayed rectifier potassium channel (IKr), encoded by the human ether-a-go-go–related gene (hERG), which is located on chromosome 7. This can induce Torsades de pointes that it is a ventricular tachycardia associated with delayed cardiac repolarization that can manifest as a prolonged QT interval on the electrocardiogram and that may cause syncope, seizure, ventricular fibrillation, and sudden death.62,63

Beyond hERG channel blockade, additional mechanisms can be involved in proarrhythmic activity of these drugs; for instance, sodium channel blockade by first generation H1A should be considered.64 Pastor et al.65 reported a case of Brugada syndrome unmasked by dimenhydrinate, hypothesising that sodium channel involvement was the basis for the observed event.65 Also antimuscarinic properties of some antihistamines may play a role in tachyarrhythmia occurrence: desloratadine, similarly to diphenhydramine, expresses high antimuscarinic activity on the heart, by resulting in a potential risk of pacemaker rhythm impairment, whereas loratadine and cetirizine showed lower affinity for cardiac muscarinic receptors.66

More recently, Yun et al.67 also suggested a molecular mechanism involved in H1A-induced cardiovascular adverse effects other than hERG channel inhibition that are associated with p21 (Cdc42/Rac)-activated kinase 1 (pak1) mRNA expression and function.67 The achievements of cellular electrophysiology and molecular genetics are shedding some light on the potential cardiotoxicity of H1A, an issue that might be crucial in specific therapeutic and toxicological settings.

Factors contributing to risk of cardiotoxicity of H1A

Importantly, it has become apparent that the QT interval, despite its obvious importance, is but 1 of several risk factors of patients using H1A with different indications and to genetic predispositions, that must be considered in order to assess a patient’s risk profile for Torsades de pointes. Numerous studies suggest that in order for a strategy to have an acceptable level of predictive accuracy and be cost effective, it must include consideration of those risk factors that are patient-specific, drug-specific, and clinical scenario-specific.4

Patient-specific risk factors for drug-induced Torsades de pointes are the same factors that are known to be associated with an increase in the QT interval, that is, female sex, bradycardia, hypokalemia, hypomagnesemia, hypocalcemia, diuretic agent use, hypothermia, and history of heart disease.68 As discussed earlier, the common feature of H1A that cause Torsades de pointes is their ability to block hERG channels and to prolong the QT interval.

Most likely, the interindividual variability in drug sensitivity and the variable influence of factors that affect each patient’s drug exposure (e.g., dose, drug metabolism, route of administration, and so forth) reduce the predictive accuracy of such ranking to levels that lack clinical utility. For example, terfenadine and astemizole have very high hERG blocking potency but are usually extensively metabolized so that it is unusual for them to reach blood levels that can cause Torsades de pointes (i.e., only when their metabolism had been inhibited or impaired).4

Analyses of case series of Torsades de pointes due to a specific drug reveal that the great majority of cases fall into certain categories of drug use that are influenced by the patients’ medical condition. Many of the patients in these reports had been successfully treated with the offending drug for extended periods of time, but developed Torsades de pointes after a change had occurred in their clinical condition or in the way it was managed. Many of these changes could have further reduced the patient’s repolarization reserve68 and caused excessive QT prolongation. The concept of repolarization reserve by Roden68 maintains that every person has a physiological cardiac repolarization reserve, which is genetically determined and compensates for any factor (e.g., drugs) that might either decrease repolarizing or increase depolarizing currents during the action potential. This last aspect is currently under intense scrutiny, with a growing awareness of the importance of genome-wide association studies, in addition to the already consolidated focus on single candidate genes (e.g. those encoding specific ion channels). So far, reduction of repolarisation reserve, due to specific polymorphysms, is supposed to be a possible mechanism predisposing to drug-induced QT prolongation.69,70

Another factor in H1A safety is the dependence on drug–drug interference at the level of metabolism, the lack of significant hepatic metabolism probably contributes to the freedom from significant cardiotoxic potential shown by some other second generation H1A, such as desloratadine, cetirizine, levocetirizine, and acrivastine.12 Beside the clinical conditions causing an increase in the H1A plasma levels well above therapeutic values, it should be emphasized that cases have also been described in which the arrhythmic episodes occurred at “therapeutic” drug concentrations.

Therefore, physicians before prescribing an H1A should be aware of the following points. If the patient has any form of cardiac disease, an H1A with minimal cardiac effects should be used. If the patient using other drugs to induced QT prolongation and Torsades de pointes such as macrolides, opiates, imidazolic compounds, antipsychotic, antimalarial or antimigraine drugs, the prescription of H1A should be cautious or avoided. If the patient shows any risk factor, such as special diet (grapefruit juice), hepatic disease, electrolytic disturbance, and use of anti-arrhythmic drugs with potential of extending QT interval, prescription of H1A should be preceded and followed by a cardiologist.

Conclusion

H1A are widely used, many of them are nonprescribed over the counter medications and well-known drugs by public. However, potentially dangerous cardiac unwanted effects of these drugs should be recognized by the general public. Patients at risk of Torsades de pointes, such as those with liver disease, congenital prolongation of the QT interval, electrolyte disturbance, arrhythmias, or other illness requiring drugs that prolong QT interval, should not be given H1A that are capable of prolonging repolarization. When patients take massive overdosage of an H1A, it is very likely that the drugs could alter cardiac conduction, contractility, and/or repolarization, depending on the agent and the magnitude of the overdose. In the next few years the perception of drug-induced cardiovascular toxicity might easily change because of the approval of safer drugs or refinements of techniques for predicting, managing, or preventing such toxicity.

Acknowledgements

None.

Conflict of interest

The authors declare that there is no conflict of interest.

References

  1. Simons FER, Simons KJ. H1 antihistamines: Current Status and Future Directions. World Allergy Organ J. 2008; 1(9):145–155.
  2. Poluzzi E, Diemberger I, De Ridder M, et al. Use of antihistamines and risk of ventricular tachyarrhythmia: a nested case–control study in five European countries from the ARITMO Project. Eur J Clin Pharmacol. 2017;73(11):1499–1510.
  3. Simons FER. Advances in H1–antihistamines. N Eng J Med. 2004;351(21):2203–2217.
  4. Woosley RL. Cardiac Actions of Antihistamines. Annu Rev Pharmacol Toxicol. 1996;36:233–252.
  5. Kalpaklioglu F, Baccioglu A. Efficacy and Safety of H1–Antihistamines: An Update. Antiinflamm Antiallergy Agents Med Chem. 2012;11(3):230–237.
  6. Taglialatela M, Timmerman H, Annunziato L. Cardiotoxic potential and CNS effects of first–generation antihistamines. Trends Pharmacol Sci. 200;21(2):52–56.
  7. Welch MJ, Meltzer EO, Simons FE. H1–antihistamines and the central nervous system. Clin Allergy Immunol. 2002;17:337–388.
  8. Galán Martínez L, Calderín Pulido AD, Fleites Vázquez A. Negative inotropic action and QT prolongation by azithromycin. Rev Cub Cardiol Cir Cardiovasc. 2017;23(4).
  9. Galán–Martínez L, Calderín–Pulido AD, Fleites–Vázquez A, et al. Ciprofloxacin, an antibiotic with cardiac actions on isolated rat hearts. J Pharm Pharmacogn Res. 2018;6(2):65–71.
  10. Schwartz P J, Woosley RL. Predicting the Unpredictable Drug–Induced QT Prolongation and Torsades de Pointes. J Am Coll Cardiol. 2016;67(13):1639–1650.
  11. Simons FER, Simons KJ. Histamine and H1–antihistamines: celebrating a century of progress. J Allergy Clin Immunol. 2011;128(6):1139–1150.
  12. Soldovieri MV, Miceli F, Taglialatela M. Cardiotoxic effects of antihistamines: from basics to clinics (. . . and back). Chem Res Toxicol. 2008;21(5):997–1004.
  13. Holgate ST, Canonica GW, Simons FER, et al. Consensus Group on New–Generation Antihistamines (CONGA): present status and recommendations. Clin Exp Allergy. 2003;33(9):1305–1324.
  14. Zareba W, Moss AJ, Rosero SZ, et al. Electrocardiographic findings in patients with diphenhydramine overdose. Am J Cardiol. 1197;80(9):1168–1173.
  15. Khalifa M, Drolet B, Daleau P, et al.Block of potassium currents in guinea pig ventricular myocytes and lengthening of cardiac repolarization in man by the histamine H1 receptor antagonist diphenhydramine. J Pharmacol Exp Ther. 1999;288(2):858–865.
  16. http://www.ema.europa.eu/ema/
  17. Vigne J, Alexandre J, Fobe F, et al. QT prolongation induced by hydroxyzine: a pharmacovigilance case report. Eur J Clin Pharmacol. 2015;71(3):379–381.
  18. Sakaguchi T, Itoh H, Ding WG, et al. Hydroxyzine, a first generation H(1)–receptor antagonist, inhibits human ether–a–go–go–related gene (HERG) current and causes syncope in a patient with the HERG mutation. J Pharmacol Sci. 2008;108(4):462–471.
  19. de Abajo FJ, Rodríguez LA. Risk of ventricular arrhythmias associated with nonsedating antihistamine drugs. Br J Clin Pharmacol. 1999;47(3):307–313.
  20. Roose SP, Dalack GW, Glassman AH, et al. Is doxepin a safer tricyclic for the heart? J Clin Psychiatry. 1991;52(8):338–341.
  21. Alter P, Tontsch D, Grimm W. Doxepin–induced torsade de pointes tachycardia. Ann Intern Med. 2001;135:384–385.
  22. Duncan RS, McPate MJ, Ridley JM, et al. Inhibition of the HERG potassium channel by the tricyclic antidepressant doxepin. Biochem Pharmacol. 2007;74(3):425–437.
  23. Pratt CM, Hertz RP, Ellis BE, et al. Risk of developing life threatening ventricular arrhythmia associated with terfenadine in comparison with over the counter antihistamines, ibuprofen and clemastine. Am J Cardiol. 1994;73(5):346–352.
  24. Ridley JM, Milnes JT, Hancox JC,. Clemastine, a conventional antihistamine, is a high potency inhibitor of the HERG K+ channel. J Mol Cell Cardiol. 2006;40(1):107–118.
  25. Hestand HE, Teske DW. Diphenhydramine hydrochloride intoxication. J Pediatr. 1997;90(6):1017−1018.
  26. Krenzelok EP, Anderson GM, Mirick M. Massive diphenhydramine overdose resulting in death. Ann Em Med. 1982;11(4):212–213.
  27. Rinder CS, D'Amato SL, Rinder HM, et al. Survival in complicated diphenhydramine overdose. Crit Care Med. 1988;16(11):1161–1162.
  28. Clark RF, Vance MV. Massive diphenhydramine poisoning resulting in a wide–complex tachycardia: successful treatment with sodium bicarbonate. Ann Emerg Med. 1992;21(3):318–321.
  29. Husain Z, Hussain K, Nair R, et al. Diphenhydramine induced QT prolongation and torsade de pointes: an uncommon effect of a common drug. Cardiol J. 2010;17(5):509–511.
  30. Al–Abri SA, Woodburn C, Olson KR, et al. Ventricular Dysrhythmias Associated with Poisoning and Drug Overdose: A 10–Year Review of Statewide Poison Control Center Data from California. Am J Cardiovasc Drugs. 2015;15(1):43–50.
  31. Farrell M, Heinrichs M, Tilelli JA. Response of life threatening dimenhydrinate intoxication to sodium bicarbonate administration. J Toxicol Clin Toxicol. 1991;29(4):527–535.
  32. Winn RE, McDonnell KP. Fatality secondary to massive overdose of dimenhydrinate. Ann Emerg Med. 1993;22(9):1481–1484.
  33. Salata JJ, Jurkiewicz NK, Wallace AA, et al. Cardiac electrophysi– ological actions of the histamine HI–receptor antagonists astemizole and terfenadine compared with chlorpheniramine and pyrilamine. Circ Res. 1995;76:110–119.
  34. Paudel G, Syed M, Kalantre S, et al. Pyrilamine–induced prolonged QT interval in adolescent with drug overdose. Pediatr Emerg Care. 2011;27(10):945–947.
  35. Chouinard G, Ghadirian AM, Jones BD. Death attributed to ventricular arrhythmia induced by thioridazine in combination with a single Contac C capsule. Can Med Assoc J. 1978;119(7):729–731.
  36. Burton BT, Rice M, Schmertzler LE. Atrioventricular block following overdose of decongestant cold medication. J Emerg Med. 1985;2(6):415–419.
  37. Hong HK, Jo SH. Block of hERG K+ Channel by Classic Histamine H1 Receptor Antagonist Chlorpheniramine. Korean J Physiol Pharmacol. 2009;13(3):215–220.
  38. Nia AM, Fuhr U, Gassanov N, et al. Torsades de pointes tachycardia induced by common cold compound medication containing chlorpheniramine. Eur J Clin Pharmacol. 2010;66(11):1173–1175.
  39. Park SJ, Kim KS, Kim EJ. Blockade of HERG K+ channel by an antihistamine drug brompheniramine requires the channel binding within the S6 residue Y652 and F656. J Appl Toxicol. 2008;28(2):104–111.
  40. Hollister LE. Hydroxyzine hydrochloride: possible adverse cardiac interactions. Psychophannacol Commun. 1975;1(1):61–65.
  41. Magera BE, Betlach ChJ, Sweatt AP, et al. Hydroxyzine intoxication in a 13–month–old child. Pediatrics. 1981;67(2):280–283.
  42. Kobayashi K, Omuro N, Takahara A. The conventional antihistamine drug cyproheptadine lacks QT–interval–prolonging action in halothane–anesthetized guinea pigs: comparison with hydroxyzine. J Pharmacol Sci. 2014;124(1):92–98.
  43. Mistraletti G, Iapichino G. Hydroxyzine and QTc interval: drugs without sin cast the first stone! Minerva Anestesiol. 2016;82(2):240–241.
  44. Schlit AF, Delaunois A, Colomar A, et al. Risk of QT prolongation and torsade de pointes associated with exposure to hydroxyzine: re–evaluation of an established drug. Pharmacol Res Perspect. 2017;5(3):e00309.
  45. Braillon A. Torsades de Pointes: The tip of the iceberg. Int J Cardiol. 2018;257:115.
  46. Kondou N, Hiasa Y, Kishi K, et al. A case of lifethreatening ventricular arrhythmias probably due to psychotropic drugs. Kokyu To Junkan. 1993;41(11):1117–1120.
  47. Jo SH, Hong HK, Chong SH, et al. H(1) antihistamine drug promethazine directly blocks hERG K(+) channel. Pharmacol Res. 2009;60(5):429–437.
  48. Sale ME, Barbey JT, Woosley RL, et al. The electrocardiographic effects of cetirizine in normal subjects. Clin Pharmacol Ther. 1994;56(3):295–301.
  49. Hekkala AM, Swan H, Väänänen H, et al. The effect of antihistamine cetirizine on ventricular repolarization in congenital long QT syndrome. J Cardiovasc Electrophysiol. 2007;18(7):691–695.
  50. Hulhoven R, Rosillon D, Letiexhe M, et al. Levocetirizine does not prolong the QT/QTc interval in healthy subjects: results from a thorough QT study. Eur J Clin Pharmacol. 2007;63(11):1011–1017.
  51. Moneret–Vautrin DA, de Chillou C, Codreanu A. Long QT syndrome in a patient with allergic rhinoconjunctivitis and auto–immune diabetes: focus on the choice of anti–H1 drugs. Eur Ann Allergy Clin Immunol. 2006;38(10):347–350.
  52. Park MH, Lee SH, Chu DH, et al. Effect of azelastine on cardiac repolarization of guinea–pig cardiomyocytes, hERG K⁺ channel, and human L–type and T–type Ca²⁺ channel. J Pharmacol Sci. 2013;20:123(1):67–77.
  53. Pesco–Koplowitz L, Hassell A, Lee P, et al. Lack of effect of erythromycin and ketoconazole on the pharmacokinetics and pharmacodynamics of steady–state intranasal levocabastine. J Clin Pharmacol. 1999;39(1):76–85.
  54. Brannan M, Reidenberg P, Radwanski E, et al. Loratadine administered concomitantly with erythromycin: pharmacokinetic and electrocardiographic evaluations. Clin Pharmacol Ther. 1995;8(3):269–278.
  55. Hey J, del Prado M, Egan RW, et al. Loratadine produces antihistamine activity without adverse CNS, ECG or cardiovascular effects in guinea pigs. Comparative studies with terfenadine and sedating antihistamines. Int Arch Allergy Immunol. 1995;107(1–3):418–419.
  56. Kreutner W, Hey JA, Chiu P, et al. Preclinical pharmacology of desloratadine, a selective and nonsedating histamine H1 receptor antagonist. 2nd communication: lack of central nervous system and cardiovascular effects. Arzneimittelforschung. 2000;50(5):441–448.
  57. Chaufour S, Caplain H, Lilienthal N, et al. Study of cardiac repolarization in healthy volunteers performed with mizolastine, a new H1–receptor antagonist. Br J Clin Pharmacol. 1999;47(5):515–520.
  58. Delauche–Cavallier MC, Chaufour S, Guérault E, et al. QT interval monitoring during clinical studies with mizolastine, a new H1 antihistamine. Clin Exp Allergy. 1999;29(3):206–211.
  59. Taglialatela M, Pannaccione A, Castaldo P, et al. Inhibition of HERG1 K(+) channels by the novel second–generation antihistamine mizolastine. Br J Pharmacol. 2000;131(6):1081–1088.
  60. Pinto YM, Van Gelder IC, Heeringga M, et al. QT lengthening and arrhythmias associated with fexofenadine. Lancet. 1999;353(451):980–981.
  61. Ando K, Nakamura Y, Hagiwara–Nagasawa M, et al. Comparison of electropharmacological effects between terfenadine and its active derivative fexofenadine using a cross–over study in halothane–anesthetized dogs to analyze variability of pharmacodynamic and pharmacokinetic profiles of terfenadine and torsadogenic risk of fexofenadine. J Toxicol Sci. 2018;43(3):183–192.
  62. Curran ME, Splawski I, Timothy KW, et al. A molecular basis for cardiac arrhythmia: HERG mutations cause long QT syndrome. Cell. 1995;80(5):795–803.
  63. Taglialatela M, Castaldo P, Pannaccione A, et al. Cardiac ion channels and antihistamines: possible mechanisms of cardiotoxicity. Clin Exp Allergy. 1999;29(3):182–189.
  64. Kuo CC, Huang RC, Lou BS. Inhibition of Na(+) current by diphenhydramine and other diphenyl compounds: molecular determinants of selective binding to the inactivated channels. Mol Pharmacol. 2000;57(1):135–143.
  65. Pastor A, Nunez A, Cantale C, et al. Asymptomatic brugada syndrome case unmasked during dimenhydrinate infusion. J Cardiovasc Electrophysiol. 2001;12(10):1192–1194.
  66. Liu H, Zheng Q, Farley JM. Antimuscarinic actions of antihistamines on the heart. J Biomed Sci. 2006;13:395–401.
  67. Yun J, Young S, Sik K, et al. P21 (Cdc42/Rac)–activated kinase 1 (pak1) is associated with cardiotoxicity induced by antihistamines. Arch Pharm Res. 2016;39(12):1644-1652.
  68. Roden DM. Predicting drug–induced QT prolongation and torsades de pointes. J Physiol. 2016;594(9):2459–2468.
  69. Niemeijer MN, van den Berg ME, Eijgelsheim M, et al. Pharmacogenetics of drug–induced QT interval prolongation: an update. Drug Saf. 2015;38(10):855–867.
  70. Roden DM. Pharmacogenetics of potassium channel blockers. Card Electrophysiol Clin. 2016;8(2):385–393.
Creative Commons Attribution License

©2018 Martínez, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.