358
Views
9
CrossRef citations to date
0
Altmetric
Reviews

A 2015 focus on preventing drug-induced arrhythmias

, , &
Pages 245-253 | Received 07 Sep 2015, Accepted 03 Nov 2015, Published online: 26 Nov 2015

References

  • Vandenberg JI, Perry MD, Perrin MJ, et al. HERG K+ channels: Structure, function, and clinical significance. Physiol Rev. 2012;92:1393–1478.
  • International conference on harmonisation; guidance on S7b nonclinical evaluation of the potential for delayed ventricular repolarization (QT interval prolongation) by human pharmaceuticals. Fed Regist. 2005;70:61133–61134.
  • International conference on harmonisation; guidance on E14 clinical evaluation of QT/QTc interval prolongation and proarrhythmic potential for non-antiarrhythmic drugs. Fed Regist. 2005;70:61134–61135.
  • Sager PT, Gintant G, Turner JR, et al. Rechanneling the cardiac proarrhythmia safety paradigm: A meeting report from the cardiac safety research consortium. Am Heart J. 2014;167:292–300.
  • Barsheshet A, Dotsenko O, Goldenberg I. Congenital long QT syndromes: Prevalence, pathophysiology and management. Paediatr Drugs. 2014;16:447–456.
  • Antzelevitch C, Nesterenko V, Shryock JC, et al. The role of late INa in development of cardiac arrhythmias. Handb Exp Pharmacol. 2014;221:137–168.
  • Zhao YT, Valdivia CR, Gurrola GB, et al. Arrhythmogenic mechanisms in ryanodine receptor channelopathies. Life Sci. 2015;58:54–58.
  • Kratz JM, Schuster D, Edtbauer M, et al. Experimentally validated hERG pharmacophore models as cardiotoxicity prediction tools. J Chem Inf Model. 2014;54:2887–2901.
  • Du-Cuny L, Chen L, Zhang S. A critical assessment of combined ligand-and structure-based approaches to hERG channel blocker modeling. J Chem Inf Model. 2011;51:2948–2960.
  • Wang S, Li Y, Xu L, et al. Recent developments in computational prediction of hERG blockage. Curr Top Med Chem. 2013;13:1317–1326.
  • Stary A, Wacker SJ, Boukharta L, et al. Toward a consensus model of the hERG potassium channel. Chem Med Chem. 2010;5:455–467.
  • Cavero I, Holzgrefe H. Comprehensive in vitro proarrhythmia assay, a novel in vitro/in silico paradigm to detect ventricular proarrhythmic liability: A visionary 21st century initiative. Expert Opin Drug Saf. 2014;13:745–758.
  • Kramer J, Obejero-Paz CA, Myatt G, et al. MICE models: Superior to the hERG model in predicting torsade de pointes. Sci Rep. 2013;3:2100.
  • O’Hara T, Virag L, Varro A, et al. Simulation of the undiseased human cardiac ventricular action potential: Model formulation and experimental validation. PLoS Comput Biol. 2011;7:e1002061.
  • Mirams GR, Davies MR, Brough SJ, et al. Prediction of thorough QT study results using action potential simulations based on ion channel screens. J Pharmacol Toxicol Methods. 2014;70:246–254.
  • Cavero I, Holzgrefe H. CiPA: Ongoing testing, future qualification procedures, and pending issues. J Pharmacol Toxicol Methods. 2015;76:27–37.
  • Wible BA, Hawryluk P, Ficker E, et al. HERG-lite: A novel comprehensive high-throughput screen for drug-induced hERG risk. J Pharmacol Toxicol Methods. 2005;52:136–145.
  • de Git KC, de Boer TP, Vos MA, et al. Cardiac ion channel trafficking defects and drugs. Pharmacol Ther. 2013;139:24–31.
  • Harkcom WT, Abbott GW. Emerging concepts in the pharmacogenomics of arrhythmias: Ion channel trafficking. Expert Rev Cardiovasc Ther. 2010;8:1161–1173.
  • Mehta A, Sequiera GL, Ramachandra CJ, et al. Re-trafficking of hERG reverses long QT syndrome 2 phenotype in human iPS-derived cardiomyocytes. Cardiovasc Res. 2014;102:497–506.
  • Carlsson L. The anaesthetised methoxamine-sensitised rabbit model of torsades de pointes. Pharmacol Ther. 2008;119:160–167.
  • Oros A, Beekman JD, Vos MA. The canine model with chronic, complete atrio-ventricular block. Pharmacol Ther. 2008;119:168–178.
  • Varkevisser R, Wijers SC, van der Heyden MA, et al. Beat-to-beat variability of repolarization as a new biomarker for proarrhythmia in vivo. Heart Rhythm. 2012;9:1718–1726.
  • Vos MA. Thorough QT studies: Are we off target? J Cardiovasc Pharmacol. 2014;64:438–439.
  • Belardinelli L, Liu G, Smith-Maxwell C, et al. A novel, potent, and selective inhibitor of cardiac late sodium current suppresses experimental arrhythmias. J Pharmacol Exp Ther. 2013;344:23–32.
  • Sanguinetti MC. HERG1 channel agonists and cardiac arrhythmia. Curr Opin Pharmacol. 2014;15:22–27.
  • Gerlach AC, Stoehr SJ, Castle NA. Pharmacological removal of human ether-a-go-go-related gene potassium channel inactivation by 3-nitro-n-(4-phenoxyphenyl) benzamide (ICA-105574). Mol Pharmacol. 2010;77:58–68.
  • Asayama M, Kurokawa J, Shirakawa K, et al. Effects of an hERG activator, ICA-105574, on electrophysiological properties of canine hearts. J Pharmacol Sci. 2013;121:1–8.
  • Meng J, Shi C, Li L, et al. Compound ICA-105574 prevents arrhythmias induced by cardiac delayed repolarization. Eur J Pharmacol. 2013;718:87–97.
  • Gessner G, Macianskiene R, Starkus JG, et al. The amiodarone derivative KB130015 activates hERG1 potassium channels via a novel mechanism. Eur J Pharmacol. 2010;632:52–59.
  • Crumb WJ Jr. Allosteric effects of erythromycin pretreatment on thioridazine block of hERG potassium channels. Br J Pharmacol. 2014;171:1668–1675.
  • Yu Z, Klaasse E, Heitman LH, et al. Allosteric modulators of the hERG K+ channel: Radioligand binding assays reveal allosteric characteristics of dofetilide analogs. Toxicol Appl Pharmacol. 2014;274:78–86.
  • Liu J, Yu Z, Van Veldhoven JP, et al. Full protection from drug-induced torsade de pointes-like arrhythmias by an allosteric IKr-modulatory mechanism in rat ventricular monolayers. abstract Europace. 2015;17(suppl 3):168–169.
  • Heist EK, Ruskin JN. Drug-induced arrhythmia. Circulation. 2010;122:1426–1435.
  • van der Heyden MA, Jonsson MK. Personalized medicine and the role of induced pluripotent stem cells. Cardiovasc Res. 2012;95:395–396.
  • Barra S, Agarwal S, Begley D, et al. Post-acute management of the acquired long QT syndrome. Postgrad Med J. 2014;90:348–358.
  • Beitland S, Platou ES, Sunde K. Drug-induced long QT syndrome and fatal arrhythmias in the intensive care unit. Acta Anaesthesiol Scand. 2014;58:266–272.
  • Coleman CI, Sood N, Chawla D, et al., Dofetilide Intravenous Magnesium Evaluation (DIME) Investigators. Intravenous magnesium sulfate enhances the ability of dofetilide to successfully cardiovert atrial fibrillation or flutter: Results of the dofetilide and intravenous magnesium evaluation. Europace. 2009;11:892–895.
  • Braam SR, Tertoolen L, van de Stolpe A, et al. Prediction of drug-induced cardiotoxicity using human embryonic stem cell-derived cardiomyocytes. Stem Cell Res. 2010;4:107–116.
  • Jonsson MK, Vos MA, Mirams GR, et al. Application of human stem cell-derived cardiomyocytes in safety pharmacology requires caution beyond hERG. J Mol Cell Cardiol. 2012;52:998–1008.
  • Karakikes I, Ameen M, Termglinchan V, et al. Human induced pluripotent stem cell-derived cardiomyocytes: Insights into molecular, cellular, and functional phenotypes. Circ Res. 2015;117:80–88.
  • Qu Y, Gao B, Fang M, et al. Human embryonic stem cell derived cardiac myocytes detect hERG-mediated repolarization effects, but not NaV1.5 induced depolarization delay. J Pharmacol Toxicol Methods. 2013;68:74–81.
  • Himmel HM. Drug-induced functional cardiotoxicity screening in stem cell-derived human and mouse cardiomyocytes: Effects of reference compounds. J Pharmacol Toxicol Methods. 2013;68:97–111.
  • Qu Y, Vargas HM. Proarrhythmia risk assessment in human induced pluripotent stem cell-derived cardiomyocytes using the MAESTRO MEA platform. Toxicol Sci. 2015;147:286–295.
  • Williams G, Mirams GR. A web portal for in-silico action potential predictions. J Pharmacol Toxicol Methods. 2015;75:10–16.
  • Moreno JD, Zhu ZI, Yang PC, et al. A computational model to predict the effects of class I anti-arrhythmic drugs on ventricular rhythms. Sci Transl Med. 2011;3:98ra83.
  • Bondarenko VE, Szigeti GP, Bett GC, et al. Computer model of action potential of mouse ventricular myocytes. Am J Physiol Heart Circ Physiol. 2004;287:H1378–1403.
  • Anderson CL, Delisle BP, Anson BD, et al. Most lqt2 mutations reduce kv11.1 (herg) current by a class 2 (trafficking-deficient) mechanism. Circulation. 2006;113:365–373.
  • Varkevisser R, Houtman MJ, Linder T, et al. Structure-activity relationships of pentamidine-affected ion channel trafficking and dofetilide mediated rescue. Br J Pharmacol. 2013;169:1322–1334.
  • Saffitz JE. Protein trafficking in cardiovascular disease: How the science has evolved and where it must go. Trends Cardiovasc Med. 2015;25:390–391.
  • Xiao S, Shaw RM. Cardiomyocyte protein trafficking: Relevance to heart disease and opportunities for therapeutic intervention. Trends Cardiovasc Med. 2015;25:379–389.
  • Behr ER, Roden D. Drug-induced arrhythmia: Pharmacogenomic prescribing? Eur Heart J. 2013;34:89–95.
  • Itoh H, Sakaguchi T, Ding WG, et al. Latent genetic backgrounds and molecular pathogenesis in drug-induced long-QT syndrome. Circ Arrhythm Electrophysiol. 2009;2:511–523.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.