Acessibilidade / Reportar erro

Cilostazol, a phosphodiesterase III inhibitor: future perspectives in atherosclerosis

CLINICAL UPDATE

Cilostazol, a phosphodiesterase III inhibitor: future prospects for atherosclerosis

Marcelo Pereira da Rosa; Gislaine Verginia Baroni; Vera Lúcia Portal

Instituto de Cardiologia do Rio Grande do Sul – FUC - Porto Alegre, RS, Brazil

Mailing Address Mailing Address: Vera Lúcia Portal Rua Padre Cacique, 222/602 90810-240 – Porto Alegre, RS E-mail: veraportal@cardiol.br

Key words: Phosphodiesterase inhibitors, atherosclerosis, cilostazol.

Pharmacological properties of cilostazol

Cilostazol was released in Japan and other Asiatic countries in 1988 and approved in the United States of America in 1999, for clinical treatment of intermittent claudication caused by POAD which is an important marker of systemic atherosclerosis.1 Study results demonstrate the superiority of cilostazol in comparison to placebo and pentoxifylline to increase walking distance without pain in 50% of the patients, and the maximum walking distance in 64% of the patients2, thus improving quality of life3.

This medication is an antithrombotic antiplatelet agent4 with vasodilation action5. There is no evidence that it prolongs bleeding time when compared to acetylsalicylic acid (ASA), clopidogrel or ticlopidine6,7, or even various combinations of these drugs8. It is a potent phosphodiesterase 3 selective inhibitor, which increases 3'-5' cyclic adenosine monophosphate (cAMP) in the thrombocytes and smooth muscle cells, and diminishes intracellular calcium which, in turn, causes cellular relaxation and vasodilation9. Cyclic AMP is one of the regulators of inflammatory and immunological reactions10.

It is metabolized via the cytochrome P450, primarily the isoenzyme CYP3A411,12, and is excreted by the kidneys. It should not be used concomitantly with ketoconazole, itraconzole, miconazole, fluconazole, fluvoxamine, fluoxetine, nefazodone, sertraline, omeprazole, erythromycin, diltiazem and quinidine; if indicated should be given in reduced dosages13.

The treatment independs of gender, age, smoking, presence of diabetes mellitus (DM), the concomitant use of betablockers or calcium antagonists14. Studies have shown that it is safe to use cilostazol for patients with acute myocardial infarction (AMI) despite the increased cardiac index, coronary flow and contractibility13.

The most common collateral effects include headache, tachycardia, palpitations, soft stools and diarrhea15. In two studies it was necessary to interrupt the use of cilostazol due to headaches in 1.7% of the patients in relation to 1.3% in those treated with placebo, while suspension for other causes was similar between the groups16,17. In the case of chronic liver disease, Child-Pugh classes B and C, it should be used with caution18. It is not advised in the case of congestive heart failure19 or for patients with left ventricle ejection fractions less than 40% 20.

The effect of cilostazol in the prevention of thrombotic complications and restenosis

The accumulation of cAMP, caused by cilostazol through the diminished phosphodiesterase 3 activity, initiates a series of events which include regulation of the tumor suppressor genes p53 and p21 and hepatocyte growth factor (HGF). The increased suppression of the p53 protein in the cellular cycle induces apoptosis in the smooth vascular muscle cells, causing an antiproliferative effect. HGF stimulates the rapid endothelial cell regeneration that inhibits neointimal formation by two mechanisms: suppressing the growth of abnormal smooth vascular muscle cells and improved endothelial function (Figure 1). These mechanisms could be responsible for preventing restenosis after coronary stenting21.


The incidence of thrombosis and restenosis after coronary stenting is still elevated, even with pharmacological devices22, and thrombotic events remain the primary cause of death after percutaneous transluminal coronary angioplasty23. In a cohort of 2229 patients who received stents coated with sirolimus or paclitaxel and were followed for nine months, the cumulative incidence of thrombosis was 1.3%, substantially higher than the rate referred in larger clinical trials (0.4% in one year for sirolimus and 0.6% in nine months for paclitaxel). The follow-up timeframe was nine months24.

Cilostazol demonstrated results similar to ticlopidine, when associated with ASA, in the inhibition of platelet aggregation induced by shear stress, after coronary intervention in patients with AMI and follow-up for three months25.

It also demonstrated analogous performance to ticlopidine in relation to the restenosis rate, both associated with ASA, in a multicenter study with 397 patients submitted to elective coronary stent placement procedures and followed for six months. In the extended follow-up of nine months, the performance of cilostazol was significantly better in relation to the number of revascularization surgeries required for target lesions26.

The first study that recognized and compared cilostazol and clopidogrel, used in conjunction with ASA, demonstrated that both had a similar effect in the prevention of thrombotic complications. No significant differences were found between cilostazol and clopidogrel for post AMI or non pharmacological stent implants for long, multiple and complex coronary lesions. The study lasted for thirty days and evaluated 689 patients27.

The effect of cilostazol on dyslipidemia

Even though the cholesterol contained in the low density lipoproteins (LDL) continues to be the main target in the treatment of dyslipidemia, the increase of HDL and reduction of triglycerides (TG) have proven to be very important in reducing cardiovascular risk28-30, particularly for people with DM31. While the isolated and fasting levels of HDL and TG are important, recent studies have shown that evaluation of triglyceride rich lipoprotein metabolism (chylomicrons and cholesterol contained in very low density lipoproteins - VLDL) during the postprandial period is more effective to verify the risk of coronary artery disease (CAD) or other atherosclerotic alterations32,33.

Generally, dietary sources of fat exceed the daily requirements and the tissues are mobilized to accumulate more nutrients than required. Zilversmit, in 1979, demonstrated that atherogenesis can occur after eating34. Other clinical studies reinforce this idea and suggest that the inefficient TG removal during the postprandial period can promote atherosclerosis35. It is important to understand that the postprandial response does not only represent the influx of TG from the diet in the circulation, but also a period when the lipoprotein composition is significantly altered36. The dimension of remodeled lipoproteins, that occurs during lipemia is directly related to the duration and size of the postprandial triglyceridemia37. Additionally, lipemia after eating promotes catabolism of HDL and its low blood concentration is associated with increased risk of CAD38. Postprandial hypertriglyceridemia stimulates the formation of small, dense LDL that are very atherogenic and increase the risk of CAD by four to six times39.

Cilostazol diminished the concentration of remaining VLDL and chylomicrons by 20%, increased HDL and diminished TG in 874 patients with POAD, during a randomized multicenter study, controlled with pentoxifylline and placebo with six months of follow-up40. In 189 individuals with POAD and without hyperlipidemia, cilostazol reduced TG by 15% and increased HDL by 9.5%, in a double blind multicenter study, controlled with placebo and twelve weeks of follow-up41. It also improved postprandial lipemia in 112 patients with type 2 DM or glucose intolerance, controlled with placebo during twelve weeks of follow-up42.

The effect of cilostazol on inflammatory markers

Atherosclerosis is characterized by endothelial lesions, adhesion of mononuclear leukocytes, migration and proliferation of smooth muscle cells, as well as extracellular matrix deposition43. It is considered as an inflammatory disease and from a pathological view point all the development stages of atherosclerotic plaque – formation, growth and complication – can be considered inflammatory responses to the endothelial lesion44. This justifies not only the aggressive handling of the modifiable risk factors but also the treatment of the causal lesion and stabilization of other lesions45.

Various adhesion molecules such as the vascular cell adhesion molecule 1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1), have been detected in atherosclerotic lesions, promoting an interaction between the endothelial surface and the circulating leukocytes, mediating their recruitment and accumulation on the intima of the blood cell wall46. VCAM-1 has an important role in mediating the selective adhesion of mononuclear leukocytes to the vascular endothelial and is a marker of early onset atherosclerosis47. MCP-1 is a chemokine which also has an important role in mediating the recruitment of monocytes in the atherosclerotic lesion48. Therefore, eliminating the expression of these adhesion molecules could be a strategy to prevent atherogenesis.

TNF-a (tumor necrosis factor alpha) is a cytokine that is implicated not only in the induction of endothelial apoptosis but also in the progression of atherosclerotic lesions49.

Interleukin-6 (IL-6) is a pro-inflammatory cytokine produced by various cell types including macrophages, lymphocytes and endothelial cells, which inhibits lipoprotein lipase, an important enzyme in the catabolism process of triglyceride rich lipoproteins and stimulates the secretion of hepatic TG50.

Cilostazol suppressed cytokine production in a mastocyte culture51. It also blocked the production and expression of MCP-1 induced by TNF-a, in culture of endothelial cells taken from a human umbilical cord52,53. It inhibited VCAM-1 in a culture of endothelial cells taken from a human umbilical cord via suppression of the nuclear transcription factor kappa B54. In rats it diminished the superoxide action, demonstrating a possible antioxidant and TNF-a effect55, it also inhibited the TNF-a in a culture of human neuroblastoma cells56.

Additionally, cilostazol increased the lipoprotein lipase activity in a culture of aorta cells from rats57, diminished IL-6 in patients submitted to revascularization procedures for POAD58 and inhibited IL-6 in a study with POAD patients controlled with pentoxifylline and placebo59.

The effect of cilostazol on nitric oxide and apoptosis

Nitric oxide (NO) provides a variety of functions to protect vessels such as vasodilation, inhibition of the migration and proliferation of smooth vascular muscle cells and stimulation of endothelial growth, preserving endothelial function; it is also involved in the regulation of coronary circulation60.

Apoptosis, or programmed cell death, is an important tissue function to maintain homeostasis through the elimination of undesired and/or harmful cells. It is associated with the development of atherosclerotic plaque and occurs with greater frequency in advanced plaques61.

Among the factors responsible for apoptosis are oxidative stress62 and the B-cell leukemia/lymphoma 2 (Bcl-2) gene family63. The Bcl-2 gene was originally identified as an oncogene of human follicular lymphoma64 and later, was suggestive of suppressing cell death by apoptosis in a variety of in vitro systems and cellular lineages, promoting cellular survival after cerebral ischemia in rats65. The Bcl2 associated X protein (BAX) is a member of the Bcl-2 family that, in opposition, promotes cell death66.

Cilostazol increased the NO expression in cell cultures67-70 and rats71, positively altered oxidative stress53,55, inhibited apoptosis induced by lipopolysaccharides; it diminished BAX gene levels and increased Bcl-2 gene levels, in cultures of endothelial cells taken from a human umbilical cord72. It also diminished cerebral stroke in association with apoptosis inhibition and oxidative cellular death in rats submitted to focal cerebral ischemia73.

Conclusion

The pharmacological treatment of atherosclerosis can diminish the rate of progression of the disease and in certain cases can also cause involution74.

Cilostazol acts as a vasodilator, antithrombotic antiplatelet agent. This drug promotes lower TG levels, increases HDL in patients with POAD41, improves postprandial lipemia in patients with DM42, increases NO expression, has a positive effect on apoptosis, prevents thrombosis after stenting and has demonstrated the ability to interfere in various stages of the atherosclerotic process.

These effects can make cilostazol an important option in the treatment of atherosclerosis. Further controlled clinical trials and studies are required to evaluate these other effects, as well as its already established role as a peripheral vasodilator.

Acknowledgements

The authors would like to thank Professors Dione Maria Detanico Busetti and Raquel Rech Lazzaron for their help in editing of this text.

Referências

1. Faxon DP, Creager MA, Smith SC, et al. Atherosclerotic vascular disease conference: executive summary: atherosclerotic vascular disease conference proceeding for healthcare professionals from a special writing group of the American Heart Association. Circulation 2004; 109: 2595-604.

2. Thompson PD, Zimet R, Forbes WP, Zhang P. Meta-analysis of results from eight randomized, placebo-controlled trials on the effect of cilostazol on patients with intermittent claudication. Am J Cardiol 2002; 90: 1314-19.

3. Regensteiner JG, Ware JE Jr, McCarthy WJ, et al. Effect of cilostazol on treadmill walking, community-based walking ability, and health-related quality of life in patients with intermittent claudication due to peripheral arterial disease: meta-analysis of six randomized controlled trials. J Am Geriatr Soc 2002; 50: 1939-46.

4. Kimura Y, Tani T, Kanbe T, Watanabe K. Effect of cilostazol on platelet aggregation and experimental thrombosis. Arzneim-Forsch / Drug Res 1985; 35: 1144-9.

5. Yasuda K, Sakuma M, Tanabe T. Hemodynamic effect of cilostazol on increasing peripheral blood flow in arteriosclerosis obliterans. Arzneim-Forsch / Drug Res 1985; 35: 1198-200.

6. Tamai Y, Takami H, Nakahata R, Ono F, Munakata A. Comparison of the effects of acetylsalicylic acid, ticlopidine and cilostazol on primary hemostasis using a quantitative bleeding time test apparatus. Haemostasis 1999; 29: 269-76.

7. Kim J, Lee K, Kim Y, Tamai Y, Nakahata R, Takami H. A randomized crossover comparative study of aspirin, cilostazol and clopidogrel in normal controls: analysis with quantitative bleeding time and platelet aggregation test. J Clin Neurosci 2004; 11: 600-2.

8. Comerota AJ. Effect on platelet function of cilostazol, clopidogrel, and aspirin, each alone or in combination. Atheroscler Suppl 2005; 6: 13-19.

9. Shrör K. The pharmacology of cilostazol. Diabetes Obes Metab 2002; 4: 14-19.

10. Katakami Y, Nakao Y, Koizumi T, Katakami N, Ogawa R, Fujita T. Regulation of tumour necrosis factor production by mouse peritoneal macrophages: the role of cellular cyclic AMP. Immunology 1988; 64: 719-24.

11. Abbas R, Chow CP, Browder NJ, et al. In vitro metabolism and interaction of cilostazol with human hepatic cytochrome P450 isoforms. Hum Exp Toxicol 2000; 19: 178-84.

12. Suri A, Forbes WP, Bramer SL. Effects of CYP3A inhibition on the metabolism of cilostazol. Clin Pharmacokinet 1999; 37: 61-8.

13. Pratt CM. Analysis of the cilostazol safety database. Am J Cardiol 2001; 87: 28-33.

14. Dawson DL. Comparative effects of cilostazol and other therapies for intermittent claudication. Am J Cardiol 2001; 87: 19-27.

15. Sorkin EM, Markham A. Cilostazol. Drugs Aging 1999; 14: 63-71.

16. Money SR, Herd JA, Isaacsohn JL, et al. Effect of cilostazol on walking distances in patients with intermittent claudication caused by peripheral vascular disease. J Vasc Surg 1998; 27: 267-74.

17. Beebe HG, Dawson DL, Cutler BS, et al. A new pharmacological treatment for intermittent claudication: results of a randomized, multicenter trial. Arch Intern Med 1999; 159: 2041-50.

18. Bramer SL, Forbes WP. Effect of hepatic impairment on the pharmacokinetics of a single dose of cilostazol. Clin Pharmacokinet 1999; 37: 25-32.

19. Regensteiner JG, Hiatt WR. Current medical therapies for patients with peripheral arterial disease: a critical review. Am J Med 2002; 112: 49-57.

20. Mukherjee D, Yadav JS. Update on peripheral vascular diseases: from smoking cessation to stenting. Cleve Clin J Med 2001; 68: 723-33.

21. Morishita R. A scientific rationale for the CREST trial results: evidence for the mechanism of action of cilostazol in restenosis. Atheroscler Suppl 2005. in press

22. Brophy JM, Belisle P, Joseph L. Evidence for use of coronary stents: a hierarchical Bayesian meta-analysis. Ann Intern Med 2003; 138: 777-86.

23. Moussa I, Di Mario C, Reimers B, Akiyama T, Tobis J, Colombo A. Subacute stent thrombosis in the era of intravascular ultrasound-guided coronary stenting without anticoagulation: frequency, predictors and clinical outcome. J Am Coll Cardiol 1997; 29: 6-12.

24. Iakovou I, Schmidt T, Bonizzoni E, et al. Incidence, predictors, and outcome of thrombosis after successful implantation of drug-eluting stents. JAMA 2005; 293: 2126-30.

25. Tanigawa T, Nishikawa M, Kitai T, et al. Increased platelet aggregability in response to shear stress in acute myocardial infarction and its inhibition by combined therapy with aspirin and cilostazol after coronary intervention. Am J Cardiol 2000; 85: 1054-9.

26. Ge J, Han H, Jiang H, et al. RACTS: a prospective randomized antiplatelet trial of cilostazol versus ticlopidine in patients undergoing coronary stenting – long-term clinical and angiographic outcome. J Cardiovasc Pharmacol 2005; 46: 162-6.

27. Lee S, Park S, Hong M, et al. Comparison of cilostazol and clopidogrel after successful coronary stenting. Am J Cardiol 2005; 95: 859-62.

28. LaRosa JC, Gotto Jr AM. Past, present, and future standards for management of dyslipidemia. Am J Med 2004; 116: 3-8.

29. Brousseau ME, Schaefer EJ. New developments in the prevention of atherosclerosis in patients with low high-density lipoprotein cholesterol. Curr Atheroscler Rep 2001; 3: 365-72.

30. Hokanson JE, Austin MA. Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies. J Cardiovasc Risk 1996; 3: 213-19.

31. Yoshino G, Hirano T, Kazumi T. Dyslipidemia in diabetes mellitus. Diab Res Clin Prac 1996; 33: 1-14.

32. Karpe F. Posprandial lipoprotein metabolism and atherosclerosis. J Intern Med 1999; 246: 341-55.

33. Eberly LE, Stamler J, Neaton JD. Relation of triglyceride levels, fasting and nonfasting, to fatal and nonfatal coronary heart disease. Arch Intern Med 2003; 163: 1077-83.

34. Zilversmit DB. Atherogenesis: a postprandial phenomenon. Circulation 1979; 60: 473-83.

35. Patsch JR, Miesenbock G, Hopferwieser T, et al. Relation of triglyceride metabolism and coronary artery disease. Studies in the postprandial state. Arterioscler Thromb 1992; 12: 1336-45.

36. Roche HM, Gibney MJ. The impact of postprandial lipemia in accelerating atherothrombosis. J Cardiovasc Risk 2000; 7: 317-24.

37. Cohn JS. Postprandial lipemia: emerging evidence for atherogenicity of remnant lipoproteins. Can J Cardiol 1998; 14: 18-27.

38. Patsch JR, Karlin JB, Scott LW, Smith LC, Gotto Jr AM. Inverse relationship between blood levels of high density lipoprotein subfraction 2 and magnitude of postprandial lipemia. Proc Natl Acad Sci USA 1983; 80: 1449-53.

39. Griffin BA, Freeman DJ, Tait GW, et al. Role of plasma triglyceride in the regulation of plasma low density lipoprotein (LDL) subfractions: relative contribution of small, dense LDL to coronary heart disease risk. Atherosclerosis 1994; 106: 241-53.

40. Wang T, Elam MB, Forbes WP, Zhong J, Nakajima K. Reduction of remnant lipoprotein cholesterol concentrations by cilostazol in patients with intermittent claudication. Atherosclerosis 2003; 171: 337-42.

41. Elam MB, Heckman J, Crouse JR, et al. Effect of the novel antiplatelet agent cilostazol on plasma lipoproteins in patients with intermittent claudication. Arterioscler Thromb Vasc Biol 1998; 18: 1942-7.

42. Ikewaki K, Mochizuki K, Iwasaki M, Nishide R, Mochizuki S, Tada N. Cilostazol, a potent phosphodiesterase type III inhibitor, selectively increases antiatherogenic high-density lipoprotein subclass LpA-I and improves postprandial lipemia in patients with type 2 diabetes mellitus. Metabolism 2002; 51: 1348-54.

43. Ross R. Atherosclerosis - an inflammatory disease. N Engl J Med 1999; 340: 115-26.

44. Libby P. Inflammation in atherosclerosis. Nature 2002; 420: 868-74.

45. Libby P, Theroux P. Pathophysiology of coronary artery disease. Circulation 2005; 111: 3481-8.

46. Bevilacqua MP, Nelson RM, Mannori G, Cecconi O. Endothelial-leukocyte adhesion molecules in human disease. Annu Rev Med 1994; 45: 361-78.

47. Otsuki M, Hashimoto K, Morimoto Y, Kishimoto T, Kasayama S. Circulating vascular cell adhesion molecule-1 (VCAM-1) in atherosclerotic NIDDM patients. Diabetes 1997; 46: 2096-101.

48. Baggiolini M, Loetscher P. Chemokines in inflammation and immunity. Immunol Today 2000; 21: 418-20.

49. Meyer JW, Holland JA, Ziegler LM, Chang MM, Beebe G, Schmitt ME. Identification of a functional leukocyte-type NADPH oxidase in human endothelial cells: a potential atherogenic source of reactive oxygen species. Endothelium 1999; 7: 11-22.

50. Van Snick J. Interleukin-6: an overview. Annu Rev Immunol 1990; 8: 253-78.

51. Shichijo M, Inagaki N, Kimata M, Serizawa I, Saito H, Nagai H. Role of cyclic 3',5'- adenosine monophosphate in the regulation of chemical mediator release and cytokine production from cultured human mast cells. J Allergy Clin Immunol 1999; 103: 421-8.

52. Nishio Y, Kashiwagi A, Takahara N, Hidaka H, Kikkawa R. Cilostazol, a cAMP phosphodiesterase inhibitor, attenuates the production of monocyte chemoattractant protein-1 in response to tumor necrosis factor-alpha in vascular endothelial cells. Horm Metab Res 1997; 29: 491-5.

53. Park SY, Lee JH, Kim YK, et al. Cilostazol prevents remnant lipoprotein particle-induced monocyte adhesion to endothelial cells by suppression of adhesion molecules and monocyte chemoattractant protein-1 expression via lectin-like receptor for oxidized low-density lipoprotein receptor activation. J Pharmacol Exp Ther 2005; 312: 1241-8.

54. Otsuki M, Saito H, Xu X, Sumitani S, Kouhara H, Kurabayashi M, Kasayama S. Cilostazol represses vascular cell adhesion molecule-1 gene transcription via inhibiting NF-kB binding to its recognition sequence. Atherosclerosis 2001; 158: 121-8.

55. Lee JH, Oh GT, Park SY, et al. Cilostazol reduces atherosclerosis by inhibition of superoxide and tumor necrosis factor-alpha formation in low-density lipoprotein receptor-null mice fed high cholesterol. J Pharmacol Exp Ther 2005; 313: 502-9.

56. Hong KW, Kim KY, Shin HK, et al. Cilostazol prevents tumor necrosis factor-a- induced cell death by suppression of phosphatase and tensin homolog deleted from chromosome 10 phosphorylation and activation of akt/cyclic AMP response element-binding protein phosphorylation. J Pharmacol Exp Ther 2003; 306: 1182- 90.

57. Tani T, Uehara K, Sudo T, Marukawa K, Yasuda Y, Kimura Y. Cilostazol, a selective type III phosphodiesterase inhibitor, decreases triglyceride and increases HDL cholesterol levels by increasing lipoprotein lipase activity in rats. Atherosclerosis 2000; 152: 299-305.

58. Nomura S, Imamura A, Okuno M, et al. Platelet-derived microparticles in patients with arteriosclerosis obliterans enhancement of high shear-induced microparticle generation by cytocines. Thromb Res 2000; 98: 257-68.

59. Lee T, Su S, Hwang J, et al. Differential lipogenic effects of cilostazol and pentoxifylline in patients with intermittent claudication: potential role for interleukin-6. Atherosclerosis 2001; 158: 471-6.

60. Russo G, Leopold JA, Loscalzo J. Vasoactive substances: nitric oxide and endothelial dysfunction in atherosclerosis. Vasc Pharmacol 2002; 38: 259-69.

61. Kockx MM. Apoptosis in the atherosclerotic plaque: quantitative and qualitative aspects. Arterioscler Thromb Vasc Biol 1998; 18: 1519-22.

62. Buttke TM, Sandstrom PA. Oxidative stress as a mediator of apoptosis. Immunol Today 1994; 15: 7-10.

63. Reed JC. Bcl-2 and the regulation of programmed cell death. J Cell Biol 1994; 124: 1-6.

64. Tsujimoto Y, Croce CM. Analysis of the structure, transcripts, and protein products of bcl-2, the gene involved in human follicular lymphoma. Proc Natl Acad Sci USA 1986; 83: 5214-18.

65. Chen J, Graham SH, Nakayama M, et al. Apoptosis repressor genes Bcl-2 and Bcl-x-long are expressed in the rat brain following global ischemia. J Cereb Blood Flow Metab 1997; 17: 2-10.

66. Xiang J, Chao DT, Korsmeyer SJ. BAX-induced cell death may not require interleukin 1ß-converting enzyme-like proteases. Proc Natl Acad Sci USA 1996; 93: 14559-63.

67. Ikeda U, Ikeda M, Kano S, Kanbe T, Shimada K. Effect of cilostazol, a cAMP phosphodiesterase inhibitor, on nitric oxide production by vascular smooth muscle cells. Eur J Pharmacol 1996; 314: 197-202.

68. Inada H, Shindo H, Tawada M, Onaya T. Cilostazol, a cyclic AMP phosphodiesterase inhibitor, stimulates nitric oxide production and sodium potassium adenosine triphosphatase activity in SH-SY5Y human neuroblastoma cells. Life Sci 1999; 65: 1413-22.

69. Ito C, Kusano E, Akimoto T, et al. Cilostazol enhances IL-1B-induced NO production and apoptosis in rat vascular smooth muscle via PKA-dependent pathway. Cell Signal 2002; 14: 625-32.

70. Omi H, Okayama N, Shimizu M, et al. Cilostazol inhibits high glucose-mediated endothelial-neutrophil adhesion by decreasing adhesion molecule expression via NO production. Microvasc Res 2004; 68: 119-25.

71. Nakamura T, Houchi H, Minami A, et al. Endothelium-dependent relaxation by cilostazol, a phosphodiesterase III inhibitor, on rat thoracic aorta. Life Sci 2001; 69: 1709-15.

72. Kim KY, Shin HK, Choi JM, Hong KW. Inhibition of lipopolysaccharide-induced apoptosis by cilostazol in human umbilical vein endothelial cells. J Pharmacol Exp Ther 2002; 300: 709-15.

73. Choi JM, Shin HK, Kim KY, Lee JH, Hong KW. Neuroprotective effect of cilostazol against focal cerebral ischemia via antiapoptotic action in rats. J Pharmacol Exp Ther 2002; 300: 787-93.

74. Lusis AJ. Atherosclerosis. Nature 2000; 407: 233-41.

Recebido em 20/12/05; revisado recebido em 02/03/06; aceito em 30/03/06.

9. Shrör K. The pharmacology of cilostazol. Diabetes Obes Metab 2002; 4: 14-19.

  • Mailing Address:
    Vera Lúcia Portal
    Rua Padre Cacique, 222/602
    90810-240 – Porto Alegre, RS
    E-mail:
  • Publication Dates

    • Publication in this collection
      06 Mar 2007
    • Date of issue
      Nov 2006
    Sociedade Brasileira de Cardiologia - SBC Avenida Marechal Câmara, 160, sala: 330, Centro, CEP: 20020-907, (21) 3478-2700 - Rio de Janeiro - RJ - Brazil, Fax: +55 21 3478-2770 - São Paulo - SP - Brazil
    E-mail: revista@cardiol.br