455
Views
12
CrossRef citations to date
0
Altmetric
Review

Emerging drugs for the treatment of angina pectoris

, &
Pages 365-376 | Received 23 May 2016, Accepted 22 Sep 2016, Published online: 06 Oct 2016

References

  • Beltrame JF, Crea F, Kaski JC, et al. The who, what, why, when, how and where of vasospastic angina. Circ J. 2016;80(2):289–298.
  • Freedman SB, Richmond DR, Kelly DT. Clinical studies of patients with coronary spasm. Am J Cardiol. 1983;52(2):67A–71A.
  • Cocco G, Jerie P. Angina pectoris in patients without flow-limiting coronary artery disease (cardiac syndrome X). A forest of a variety of trees. Cardiol J. 2015;22(6):605–612.
  • Ahn JH, Kim SM, Park SJ, et al. Coronary microvascular dysfunction as a mechanism of angina in severe as: prospective adenosine-stress CMR study. J Am Coll Cardiol. 2016;67(12):1412–1422.
  • Nakahashi T, Naka M, Shiotani I, et al. Vasospastic angina pectoris associated with apical hypertrophic cardiomyopathy. Intern Med. 1995;34(5):436–440.
  • Suzuki N, Seto S, Koide Y, et al. Coexistence of familial hypertrophic cardiomyopathy and vasospastic angina pectoris in two brothers. Jpn Heart J. 2003;44(5):775–782.
  • Di Fiore DP, Beltrame JF. Chest pain in patients with ‘normal angiography’: could it be cardiac? Int J Evid Based Healthc. 2013;11(1):56–68.
  • Munzel T, Steven S, Daiber A. Organic nitrates: update on mechanisms underlying vasodilation, tolerance and endothelial dysfunction. Vascul Pharmacol. 2014;63(3):105–113.
  • Cohn JN, Archibald DG, Ziesche S, et al. Effect of vasodilator therapy on mortality in chronic congestive heart failure. Results of a veterans administration cooperative study. N Engl J Med. 1986;314(24):1547–1552.
  • Taylor AL, Ziesche S, Yancy C, et al. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med. 2004;351(20):2049–2057.
  • Boden WE, O’Rourke RA, Teo KK, et al. Optimal medical therapy with or without PCI for stable coronary disease. N Eng J Med. 2007;356(15):1503–1516.
  • Cheng DC, Jiang MT, Asokumar B, et al. Comparison of nifedipine and metoprolol on collateral coronary blood flow in a swine model of chronic coronary obstruction and acute ischaemia during isoflurane anaesthesia. Can J Anaesth. 1996;43(2):160–168.
  • Yokoyama M, Koizumi T, Fujitani K, et al. Adverse response to nifedipine in unstable angina pectoris. Chest. 1982;81(5):646–648.
  • Kezerashvili A, Marzo K, De Leon J. Beta blocker use after acute myocardial infarction in the patient with normal systolic function: when is it “ok” to discontinue? Curr Cardiol Rev. 2012;8(1):77–84.
  • Poirier L, Tobe SW. Contemporary use of beta-blockers: clinical relevance of subclassification. Can J Cardiol. 2014;30(5 Suppl):S9–S15.
  • Randle PJ, Garland PB, Hales CN, et al. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1(7285):785–789.
  • Beadle RM, Frenneaux M. Modification of myocardial substrate utilisation: a new therapeutic paradigm in cardiovascular disease. Heart. 2010;96(11):824–830.
  • Yusuf S, Sleight P, Pogue J, et al. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The heart outcomes prevention evaluation study investigators. N Engl J Med. 2000;342(3):145–153.
  • Chow BS, Allen TJ. Angiotensin II type 2 receptor (AT2R) in renal and cardiovascular disease. Clin Sci (Lond). 2016;130(15):1307–1326.
  • Sverdlov AL, Chan WPA, Procter NEK, et al. Reciprocal regulation of NO signaling and TXNIP expression in humans: impact of aging and ramipril therapy. Int J Cardiol. 2013;168(5):4624–4630.
  • Simko F, Simko J, Fabryova M. ACE-inhibition and angiotensin II receptor blockers in chronic heart failure: pathophysiological consideration of the unresolved battle. Cardiovasc Drugs Ther. 2003;17(3):287–290.
  • Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomised trial–the losartan heart failure survival study ELITE II. Lancet. 2000;355(9215):1582–1587.
  • Dickstein K, Kjekshus J. Effects of losartan and captopril on mortality and morbidity in high-risk patients after acute myocardial infarction: the OPTIMAAL randomised trial. Optimal trial in myocardial infarction with angiotensin II antagonist losartan. Lancet. 2002;360(9335):752–760.
  • Bussmann WD, Goerke S, Schneider W, et al. [Angiotensin-converting enzyme inhibitors in angina pectoris]. Dtsch Med Wochenschr. 1988;113(14):548–550.
  • Antonopoulos AS, Margaritis M, Shirodaria C, et al. Translating the effects of statins: from redox regulation to suppression of vascular wall inflammation. Thromb Haemost. 2012;108(5):840–848.
  • Deanfield JE, Sellier P, Thaulow E, et al. Potent anti-ischaemic effects of statins in chronic stable angina: incremental benefit beyond lipid lowering? Eur Heart J. 2010;31(21):2650–2659.
  • Lee L, Horowitz J, Frenneaux M. Metabolic manipulation in ischaemic heart disease, a novel approach to treatment. Eur Heart J. 2004;25(8):634–641.
  • Ashrafian H, Horowitz JD, Frenneaux MP. Perhexiline. Cardiovasc Drug Rev. 2007;25(1):76–97.
  • Forbes GB, Rake MO, Taylor DJ. Liver damage due to perhexiline maleate. J Clin Pathol. 1979;32(12):1282–1285.
  • Lewis D, Wainwright HC, Kew MC, et al. Liver damage associated with perhexiline maleate. Gut. 1979;20(3):186–189.
  • Pieterse AS, Rowland R, Dunn D. Perhexiline maleate induced cirrhosis. Pathology. 1983;15(2):201–203.
  • Goble AJ, Horowitz JD. Perhexilene neuropathy: a report of two cases. Aust N Z J Med. 1984;14(3):279.
  • Horowitz JD, Sia ST, Macdonald PS, et al. Perhexiline maleate treatment for severe angina pectoris–correlations with pharmacokinetics. Int J Cardiol. 1986;13(2):219–229.
  • Phuong, H, Choi BY, Chong CR, et al. Can perhexiline be utilized without long-term toxicity? A clinical practice audit. Ther Drug Monit. 2016; 38 (1): 73–8.
  • Kennedy JA, Unger SA, Horowitz JD. Inhibition of carnitine palmitoyltransferase-1 in rat heart and liver by perhexiline and amiodarone. Biochem Pharmacol. 1996;52(2):273–280.
  • Kennedy JA, Beck-Oldach K, McFadden-Lewis K, et al. Effect of the anti-anginal agent, perhexiline, on neutrophil, valvular and vascular superoxide formation. Eur J Pharmacol. 2006;531(1–3):13–19.
  • Ngo, D.T, Drury NE, Pagano D, et al. How does perhexiline maleate modulate myocardial energetics and ameliorate redox stress? Circulation. 2011;124:A14461.
  • Lee L, Campbell R, Scheuermann-Freestone M, et al. Metabolic modulation with perhexiline in chronic heart failure: a randomized, controlled trial of short-term use of a novel treatment. Circulation. 2005;112(21):3280–3288.
  • Abozguia K, Elliott P, McKenna W, et al. Metabolic modulator perhexiline corrects energy deficiency and improves exercise capacity in symptomatic hypertrophic cardiomyopathy. Circulation. 2010;122(16):1562–1569.
  • Unger SA, Robinson MA, Horowitz JD. Perhexiline improves symptomatic status in elderly patients with severe aortic stenosis. Aust NZ J Med. 1997;27(1):24–28.
  • Australian medicines handbook. Adelaide: Australian Medicines Handbook Pty Ltd; 2015.
  • Gould BJ, Amoah AG, Parke DV. Stereoselective pharmacokinetics of perhexiline. Xenobiotica. 1986;16(5):491–502.
  • Licari G, Sallustio BC, Somogyi AA, et al. The enantiomers of the myocardial metabolic agent perhexiline display divergent effets on hepatic energy metabolism and peripheral neural function in rats. Global Heart. 2014;9(Supplement 1):e272.
  • Chong CR, Drury NE, Licari G, et al. Stereoselective handling of perhexiline: implications regarding accumulation within the human myocardium. Eur J Clin Pharmacol. 2015;71(12):1485–1491.
  • Kantor PF, Lucien A, Kozak R, et al. The antianginal drug trimetazidine shifts cardiac energy metabolism from fatty acid oxidation to glucose oxidation by inhibiting mitochondrial long-chain 3-ketoacyl coenzyme A thiolase. Circ Res. 2000;86(5):580–588.
  • Kennedy JA, Horowitz JD. Effect of trimetazidine on carnitine palmitoyltransferase-1 in the rat heart. Cardiovasc Drugs Ther. 1998;12(4):359–363.
  • Peng S, Zhao M, Wan J, et al. The efficacy of trimetazidine on stable angina pectoris: a meta-analysis of randomized clinical trials. Int J Cardiol. 2014;177(3):780–785.
  • Fragasso G, Perseghin G, De Cobelli F, et al. Effects of metabolic modulation by trimetazidine on left ventricular function and phosphocreatine/adenosine triphosphate ratio in patients with heart failure. Eur Heart J. 2006;27(8):942–948.
  • Tuunanen H, Engblom E, Naum A, et al. Trimetazidine, a metabolic modulator, has cardiac and extracardiac benefits in idiopathic dilated cardiomyopathy. Circulation. 2008;118(12):1250–1258.
  • Danchin N, Marzilli M, Parkhomenko A, et al. Efficacy comparison of trimetazidine with therapeutic alternatives in stable angina pectoris: a network meta-analysis. Cardiology. 2011;120(2):59–72.
  • Rosano GMC, Vitale C, Volterrani M. Pharmacological management of chronic stable angina: focus on ranolazine. Cardiovasc Drugs Ther. 2016.
  • Wimmer NJ, Stone PH. Anti-anginal and anti-ischemic effects of late sodium current inhibition. Cardiovasc Drugs Ther. 2013;27(1):69–77.
  • Clarke B, Wyatt KM, McCormack JG. Ranolazine increases active pyruvate dehydrogenase in perfused normoxic rat hearts: evidence for an indirect mechanism. J Mol Cell Cardiol. 1996;28(2):341–350.
  • Stone PH, Gratsiansky NA, Blokhin A, et al. Antianginal efficacy of ranolazine when added to treatment with amlodipine: the ERICA (Efficacy of Ranolazine in Chronic Angina) trial. J Am Coll Cardiol. 2006;48(3):566–575.
  • Kosiborod M, Arnold SV, Spertus JA, et al. Evaluation of ranolazine in patients with type 2 diabetes mellitus and chronic stable angina: results from the TERISA randomized clinical trial (Type 2 Diabetes Evaluation of Ranolazine in Subjects With Chronic Stable Angina). J Am Coll Cardiol. 2013;61(20):2038–2045.
  • Willoughby SR, Chirkov YY, Kennedy JA, et al. Inhibition of long-chain fatty acid metabolism does not affect platelet aggregation responses. Eur J Pharmacol. 1998;356(2–3):207–213.
  • Schmidt-Schweda S, Holubarsch C. First clinical trial with etomoxir in patients with chronic congestive heart failure. Clin Sci (Lond). 2000;99(1):27–35.
  • Turcani M, Rupp H. Modification of left ventricular hypertrophy by chronic etomixir treatment. Br J Pharmacol. 1999;126(2):501–507.
  • Cabrero A, Merlos M, Laguna JC, et al. Down-regulation of acyl-CoA oxidase gene expression and increased NF-kappaB activity in etomoxir-induced cardiac hypertrophy. J Lipid Res. 2003;44(2):388–398.
  • Gunther J, Wagner K, Theres H, et al. Myocardial contractility after infarction and carnitine palmitoyltransferase I inhibition in rats. Eur J Pharmacol. 2000;406(1):123–126.
  • Dhalla NS, Golfman L, Liu X, et al. Subcellular remodeling and heart dysfunction in cardiac hypertrophy due to pressure overload. Ann NY Acad Sci. 1999;874:100–110.
  • Sodi-Pallares D, Testelli MR, Fishleder BL, et al. Effects of an intravenous infusion of a potassium-glucose-insulin solution on the electrocardiographic signs of myocardial infarction. A preliminary clinical report. Am J Cardiol. 1962;9:166–181.
  • Kido Y, Nakae J, Accili D. Clinical review 125: the insulin receptor and its cellular targets. J Clin Endocrinol Metab. 2001;86(3):972–979.
  • Bonds DE. DIGAMI 1: 20 years later. Lancet Diabetes Endocrinol. 2014;2(8):603–604.
  • Malmberg K, Rydén L, Efendic S, et al. Randomized trial of insulin-glucose infusion followed by subcutaneous insulin treatment in diabetic patients with acute myocardial infarction (DIGAMI study): effects on mortality at 1 year. J Am Coll Cardiol. 1995;26(1):57–65.
  • Worthley MI, Holmes AS, Willoughby SR, et al. The deleterious effects of hyperglycemia on platelet function in diabetic patients with acute coronary syndromes mediation by superoxide production, resolution with intensive insulin administration. J Am Coll Cardiol. 2007;49(3):304–310.
  • Mellbin LG, Malmberg K, Norhammar A, et al. Prognostic implications of glucose-lowering treatment in patients with acute myocardial infarction and diabetes: experiences from an extended follow-up of the Diabetes Mellitus Insulin-Glucose Infusion in Acute Myocardial Infarction (DIGAMI) 2 study. Diabetologia. 2011;54(6):1308–1317.
  • Jin PY, Zhang H-S, Guo X-Y, et al. Glucose-insulin-potassium therapy in patients with acute coronary syndrome: a meta-analysis of randomized controlled trials. BMC Cardiovasc Disord. 2014;14:169.
  • Wargovich TJ, MacDonald RG, Hill JA, et al. Myocardial metabolic and hemodynamic effects of dichloroacetate in coronary artery disease. Am J Cardiol. 1988;61(1):65–70.
  • McVeigh JJ, Lopaschuk GD. Dichloroacetate stimulation of glucose oxidation improves recovery of ischemic rat hearts. Am J Physiol. 1990;259(4 Pt 2):H1079–85.
  • Wambolt RB, Lopaschuk GD, Brownsey RW, et al. Dichloroacetate improves postischemic function of hypertrophied rat hearts. J Am Coll Cardiol. 2000;36(4):1378–1385.
  • Stacpoole PW, Harman EM, Curry SH, et al. Treatment of lactic acidosis with dichloroacetate. N Engl J Med. 1983;309(7):390–396.
  • Stacpoole PW, Wright EC, Baumgartner TG, et al. A controlled clinical trial of dichloroacetate for treatment of lactic acidosis in adults. The dichloroacetate-lactic acidosis study group. N Engl J Med. 1992;327(22):1564–1569.
  • Kaufmann P, Engelstad K, Wei Y, et al. Dichloroacetate causes toxic neuropathy in MELAS: a randomized, controlled clinical trial. Neurology. 2006;66(3):324–330.
  • Gilde AJ, Van Bilsen M. Peroxisome proliferator-activated receptors (PPARS): regulators of gene expression in heart and skeletal muscle. Acta Physiol Scand. 2003;178(4):425–434.
  • Nikolaidis LA, Levine TB. Peroxisome proliferator activator receptors (PPAR), insulin resistance, and cardiomyopathy: friends or foes for the diabetic patient with heart failure? Cardiol Rev. 2004;12(3):158–170.
  • Sharma AM, Staels B. Review: peroxisome proliferator-activated receptor gamma and adipose tissue–understanding obesity-related changes in regulation of lipid and glucose metabolism. J Clin Endocrinol Metab. 2007;92(2):386–395.
  • Varas-Lorenzo C, Margulis AV, Pladevall M, et al. The risk of heart failure associated with the use of noninsulin blood glucose-lowering drugs: systematic review and meta-analysis of published observational studies. BMC Cardiovasc Disord. 2014;14:129.
  • Morita S, Mizuno Y, Harada E, et al. Pioglitazone, a peroxisome proliferator-activated receptor gamma activator, suppresses coronary spasm. Coron Artery Dis. 2014;25(8):671–677.
  • Asakawa M, Takano H, Nagai T, et al. Peroxisome proliferator-activated receptor gamma plays a critical role in inhibition of cardiac hypertrophy in vitro and in vivo. Circulation. 2002;105(10):1240–1246.
  • Noman A, Ang DSC, Ogston S, et al. Effect of high-dose allopurinol on exercise in patients with chronic stable angina: a randomised, placebo controlled crossover trial. Lancet. 2010;375(9732):2161–2167.
  • Saugstad OD. Role of xanthine oxidase and its inhibitor in hypoxia: reoxygenation injury. Pediatrics. 1996;98(1):103–107.
  • Rajaratnam R, Brieger DB, Hawkins R, et al. Attenuation of anti-ischemic efficacy during chronic therapy with nicorandil in patients with stable angina pectoris. Am J Cardiol. 1999;83(7):1120–4, A9.
  • Group IS. Effect of nicorandil on coronary events in patients with stable angina: the Impact Of Nicorandil in Angina (IONA) randomised trial. Lancet. 2002;359(9314):1269–1275.
  • Belsey J, Savelieva I, Mugelli A, et al. Relative efficacy of antianginal drugs used as add-on therapy in patients with stable angina: a systematic review and meta-analysis. Eur J Prev Cardiol. 2015;22(7):837–848.
  • Chen J, Cha-Molstad H, Szabo A, et al. Diabetes induces and calcium channel blockers prevent cardiac expression of proapoptotic thioredoxin-interacting protein. Am J Physiol Endocrinol Metab. 2009;296(5):E1133–9.
  • Park SY, Shi X, Pang J, et al. Thioredoxin-interacting protein mediates sustained VEGFR2 signaling in endothelial cells required for angiogenesis. Arterioscler Thromb Vasc Biol. 2013;33(4):737–743.
  • Vicari RM, Chaitman B, Keefe D, et al. Efficacy and safety of fasudil in patients with stable angina: a double-blind, placebo-controlled, phase 2 trial. J Am Coll Cardiol. 2005;46(10):1803–1811.
  • Ussher JR, Lopaschuk GD. Targeting malonyl CoA inhibition of mitochondrial fatty acid uptake as an approach to treat cardiac ischemia/reperfusion. Basic Res Cardiol. 2009;104(2):203–210.
  • Stanley WC, Morgan EE, Huang H, et al. Malonyl-CoA decarboxylase inhibition suppresses fatty acid oxidation and reduces lactate production during demand-induced ischemia. Am J Physiol Heart Circ Physiol. 2005;289(6):H2304–9.
  • Samokhvalov V, Ussher JR, Fillmore N, et al. Inhibition of malonyl-CoA decarboxylase reduces the inflammatory response associated with insulin resistance. Am J Physiol Endocrinol Metab. 2012;303(12):E1459–68.
  • Ritchie RH, Zeitz CJ, Wuttke RD, et al. Attenuation of the negative inotropic effects of metoprolol at short cycle lengths in humanscomparison with sotalol and verapamil. J Am Coll Cardiol. 2006;48(6):1234–1241.
  • Wiviott SD, Braunwald E, McCabe CH, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. New Engl J Med. 2007;357(20):2001–2015.
  • Mohri M, Shimokawa H, Hirakawa Y, et al. Rho-kinase inhibition with intracoronary fasudil prevents myocardial ischemia in patients with coronary microvascular spasm. J Am Coll Cardiol. 2003;41(1):15–19.
  • Kim, J.H, Shin ES, Lee JH, et al. A randomized multicenter double-blind placebo-controlled trial to evaluate the efficacy and safety of cilostazol in patients with vasospastic angina (STELLA trial). Eur Heart J. 2014;34(suppl 1): 648–649.
  • Yoo S-Y, Song S-G, Lee J-H, et al. Efficacy of cilostazol on uncontrolled coronary vasospastic angina: a pilot study. Cardiovascular Therapeutics. 2013;31(3):179–185.
  • Cole PL, Beamer AD, McGowan N, et al. Efficacy and safety of perhexiline maleate in refractory angina. A double-blind placebo-controlled clinical trial of a novel antianginal agent. Circulation. 1990;81(4):1260–1270.
  • Gutierrez JA, Karwatowska-Prokopczuk E, Murphy SA, et al. Effects of ranolazine in patients with chronic angina in patients with and without percutaneous coronary intervention for acute coronary syndrome: observations from the MERLIN-TIMI 36 trial. Clin Cardiol. 2015;38(8):469–475.
  • Marti Masso JF, Martí I, Carrera N, et al. Trimetazidine induces parkinsonism, gait disorders and tremor. Therapie. 2005;60(4):419–422.
  • Masmoudi K, Masson H, Gras V, et al. Extrapyramidal adverse drug reactions associated with trimetazidine: a series of 21 cases. Fundam Clin Pharmacol. 2012;26(2):198–203.
  • Bondon-Guitton E, Perez-Lloret S, Bagheri H, et al. Drug-induced parkinsonism: a review of 17 years’ experience in a regional pharmacovigilance center in France. Mov Disord. 2011;26(12):2226–2231.
  • Beltrame JF, Turner SP, Leslie SL, et al. The angiographic and clinical benefits of mibefradil in the coronary slow flow phenomenon. J Am Coll Cardiol. 2004;44(1):57–62.
  • Veronese ML, Gillen LP, Dorval EP, et al. Effect of mibefradil on CYP3A4 in vivo. J Clin Pharmacol. 2003;43(10):1091–1100.

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.