327
Views
11
CrossRef citations to date
0
Altmetric
Review

The pharmacodynamics of antiplatelet compounds in thrombosis treatment

, , , &
Pages 615-632 | Received 02 Sep 2015, Accepted 05 Apr 2016, Published online: 26 Apr 2016

References

  • Vane JR, Botting M. Aspirin and other salicylates. London: Chapman & Hall Medical; 1992; p. 548–575.
  • Mellion BT, Ignarro LJ, Ohlstein EH, et al. Evidence for the inhibitory role of guanosine 3¢, 5¢-monophosphate in ADP-induced human platelet aggregation in the presence of nitric oxide and related vasodilators. Blood. 1981;57:946–955.
  • Radomski MW, Palmer RM, Moncada S. Comparative pharmacology of endothelium-derived relaxing factor, nitric oxide and prostacyclin in platelets. Br J Pharmacol. 1987;92:181–187.
  • Chirkov YY, Horowitz JD. Impaired tissue responsiveness to organic nitrates and nitric oxide: a new therapeutic frontier? Pharmacol Ther. 2007;116:287–305.
  • vanGeet C, Izzi B, Labarque V, et al. Human platelet pathology related to defects in the G-protein signaling cascade. J Thromb Haemost. 2009;7(Suppl. 1):282–286.
  • Shinohara Y, Katayama Y, Uchiyama S, et al. Cilostazol for prevention of secondary stroke (CSPS 2): an aspirin-controlled, double-blind, randomised non-inferiority trial. Lancet Neurol. 2010;9:959–968.
  • Rukoyatkina N, Walter U, Friebe A, et al. Differentiation of cGMP-dependent and -independent nitric oxide effects on platelet apoptosis and reactive oxygen species production using platelets lacking soluble guanylyl cyclase. Thromb Haemost. 2011;106:922–933.
  • Schwarz UR, Walter U, Eigenthaler M. Taming platelets with cyclic nucleotides. Biochem Pharmacol. 2001;62:1153–1561.
  • Awtry EA, Loscalzo J. Cardiovascular drugs – aspirin. Circulation. 2000;101:1206–1218.
  • Patrono C, Coller B, FitzGerald GA, et al. Platelet-active drugs: the relationships among dose, effectiveness, and side effects. The Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy. Chest. 2004;126:234–264.
  • Palomo I, Toro C, Alarcón M. The role of platelets in the pathophysiology of atherosclerosis (Review). Mol Med Rep. 2008;1(2):179–184.
  • Meadows TA, Bhatt DL. Clinical aspects of platelet inhibitors and thrombus formation. Circ Res. 2007;100(9):1261–1275.
  • Alli O, Smith C, Hoffman M, et al. Incidence, predictors, and outcomes of gastrointestinal bleeding in patients on dual antiplatelet therapy with aspirin and clopidogrel. J Clin Gastroenterol. 2011;45(5):410–414.
  • Beijing institute of pharmaceutical industry. Research on the active ingredients of Ligustrium wallichii franchat. III. Clinical (218 cases) use of ligustrazine in the treatment of hypoxemic cerebral vascular diseases. Chin Med J. 1997;8:467–469.
  • Liu SY, Sylvester DM. Antithrombotic/antiplatelet activity of tetramethylpyrazine. Thromb Res. 1990;58:129–140.
  • Ho WKK, Wen HL, Lee CM. Tetramethylpyrazine for treatment of experimentally induced stroke in Mongolian gerbils. Stroke. 1989;20:96–99.
  • Sheu JR, Kan YC, Hung WC, et al. Mechanisms involved in the antiplatelet activity of tetramethylpyrazine in human platelets. Thromb Res. 1997;88:259–270.
  • Feng J, Li F, Zhao Y, et al. Brain pharmacokinetics of tetramethylpyrazine after intranasal and intravenous administration in awake rats. Int J Pharm. 2009;375(1–2):55–60.
  • Huang X, Ren P, Wen AD, et al. Pharmacokinetics of traditional Chinese syndrome and recipe: a hypothesis and its verification (I). World J Gastroenterol. 2000;6(3):384–391.
  • Meng D, Lu H, Huang S, et al. Comparative pharmacokinetics of tetramethylpyrazine phosphate in rat plasma and extracellular fluid of brain after intranasal, intragastric and intravenous administration. Acta Pharm Sin B. 2014;4(1):74–78.
  • Miller CO, Skoog F, Von Saltza MH, et al. Kinetin, a cell division factor from deoxyribonucleic acid. J Am ChemSoc. 1955;77:1392.
  • Minorsky PV. Kinetin: the elixir of life? Plant Physiol. 2003;132:1135–1136.
  • Hsiao G, Shen MY, Lin KH, et al. Inhibitory activity of kinetin on free radical formation of activated platelets in vitro and on thrombus formation in vivo. Eur J Pharmacol. 2003;465(3):281–287.
  • Sheu JR, Teng CM, Huang TF. Triflavin, an RGD containing antiplatelet peptide, binds to GP IIIa of ADP stimulated platelets. Biochem Biophys Res Commun. 1992;189:1236–1242.
  • Griffaut B, Bos R, Maurizis JC, et al. Cytotoxic effects of kinetin riboside on mouse, human and plant tumor cells. Int J Biol Macromol. 2004;34(4):271–275.
  • Sheu JR, Lee CR, Lin CC, et al. The antiplatelet activity of PMC, a potent R-tocopherol analogue, is mediated through inhibition of cyclo-oxygenase. Br J Pharmacol. 1999;127:1206–1212.
  • Nagasawa T, Tabata N, Ito Y, et al. Dietary G-rutin suppresses glycation in tissue proteins of streptozotocin-induced diabetic rats. Mol Cell Biochem. 2003;252:141–147.
  • Sheu JR, Lin CH, Peng CH, et al. Triflavin, an Arg-Gly-Asp-containing peptide, inhibits the adhesion of tumor cells to matrix proteins via binding to multiple integrin receptors expressed on human hepatoma cells. Proc Soc Exp Biol Med. 1996;213:71–79.
  • Chen WM, Jin M, Wu W. Experimental study on inhibitory effect of rutin against platelet activation induced by platelet activating factor in rabbits. Zhongguo Zhong Xi Yi Jie He ZaZhi. 2002;22(4):283–285.
  • Choi JH, Kim DW, Park SE, et al. Anti-thrombotic effect of rutin isolated from Dendropanaxmorbifera Leveille. J Biosci Bioeng. 2015;120(2):181–186.
  • Sheu JR, Hsiao G, Chou PH, et al. Mechanisms involved in the antiplatelet activity of rutin, a glycoside of the flavonol quercetin, in human platelets. J Agric Food Chem. 2004;52(14):4414–4418.
  • Cermak R, Wolffram S. Comment on lymphatic absorption of quercetin and rutin in rat and their pharmacokinetics in systemic plasma. J Agric Food Chem. 2010;58(1):546–551.
  • Voutilainen S, Nurmi T, Mursu J, et al. Carotenoids and cardiovascular health. Am J Clin Nutr. 2006;83:1265–1271.
  • Maiani G, Periago Castón MJ, Catasta G, et al. Carotenoids: actual knowledge on food sources, intakes, stability and bioavailability and their protective role in humans. Mol Nutr Food Res. 2009;53:194–218.
  • Stahl W, Sies H. Antioxidant activity of carotenoids. Mol Aspects Med. 2003;24:345–351.
  • Bosland M, Özten-Kandas N. Chemoprevention of prostate cancer: natural compounds, antiandrogens, and antioxidants - In vivo evidence. J Carcinog. 2011;10:27.
  • Sonmez M, Turk G, Ceribasi AO, et al. Attenuating effect of lycopeneandellagic acid on 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced spermiotoxic-ity and testicular apoptosis. Drug ChemToxicol. 2011;34:347–356.
  • Dahan K, Fennal M, Kumar NB. Lycopene in the prevention of prostate cancer. J Soc Integr Oncol. 2008;6:29–36.
  • Hsiao G, Wang Y, Tzu NH, et al. Inhibitory effects of lycopene on in vitro platelet activation and in vivo prevention of thrombus formation. J Lab Clin Med. 2005;146(4):216–226.
  • Sheu JR, Lee CR, Hsiao G, et al. Comparison of the relative activities of α-tocopherol and PMC on platelet aggregation and antioxidative activity. Life Sci. 1999;65:197–206.
  • Faisal W, O’Driscoll CM, Griffin BT. Bioavailability of lycopene in the rat: the role of intestinal lymphatic transport. J Pharm Pharmacol. 2010;62(3):323–331.
  • Romay C, González R, Ledón N, et al. CPhycocyanin: a biliprotein with antioxidant, anti-inflammatory and neuroprotective effects. Curr Protein Pept Sci. 2003;4:207–216.
  • Romay C, Delgado R, Remirez D, et al. Effects of phycocyanin extract on tumor necrosis factor-alpha and nitrite levels in serum of mice treated with endotoxin. Arzneimittel-Forschung. 2001;51:733–736.
  • Chiu HF, Yang SP, Kuo YL, et al. Mechanisms involved in the antiplatelet effect of C-phycocyanin. Br J Nutr. 2006;95(2):435–440.
  • Hsiao G, Chou PH, Shen MY, et al. C-phycocyanin, a very potent and novel platelet aggregation inhibitor from Spirulina platensis. J Agric Food Chem. 2005;53(20):7734–7740.
  • Akyol S, Ozturk G, Ginis Z, et al. In vivo and in vitro antineoplastic actions of caffeic acid phenethyl ester (CAPE): therapeutic perspectives. Nutr Cancer. 2013;65:515–526.
  • Hsiao G, Lee JJ, Lin KH, et al. Characterization of a novel and potent collagen antagonist, caffeic acid phenethyl ester, in human platelets: in vitro and in vivo studies. Cardiovasc Res. 2007;75(4):782–792.
  • Li Z, Xi X, Gu M, et al. A stimulatory role for cGMP-dependent protein kinase in platelet activation. Cell. 2003;112:77–86.
  • Rosado JA, Sage SO. The ERK cascade, a new pathway involved in the activation of store-mediated calcium entry in human platelets. Trends Cardiovasc Med. 2002;12:229–234.
  • Hughes PE, Renshaw MW, Pfaff M, et al. Suppression of integrin activation: a novel function of a Ras/ Raf-initiated MAP kinase pathway. Cell. 1997;88:521–530.
  • Saklatvala J, Rawlinson L, Waller RJ, et al. Role for p38 mitogen-activated protein kinase in platelet aggregation caused by collagen or a thromboxane analogue. J Biol Chem. 1996;271:6586–6589.
  • Coulon L, Calzada C, Moulin P, et al. Activation of p38 mitogen-activated protein kinase/cytosolic phospholipase A2 cascade in hydroperoxide-stressed platelets. Free Radic Biol Med. 2003;35:616–625.
  • Franke TF, Yang SI, Chan TO, et al. The protein kinase encoded by the Akt proto-oncogene is a target of the PDGF-activated phosphatidylinositol 3-kinase. Cell. 1995;81:727–736.
  • Iuliano L, Colavita AR, Leo R, et al. Oxygen free radicals and platelet activation. Free Radic Biol Med. 1997;22:999–1006.
  • Leo R, Ghiselli A, Iuliano L, et al. Detection of hydroxyl radicals by the salicylate bis-hydroxylation during arachidonic acid-dependent platelet activation. Thromb Haemost. 1995;73:A347.
  • Waldmann R, Nieberding M, Walter U. Vasodilator-stimulated protein phosphorylation in platelets is mediated by cAMP- and cGMP-dependent protein kinases. Eur J Biochem. 1987;167:441–448.
  • Halbrügge M, Walter U. Purification of a vasodilator-regulated phosphoprotein from human platelets. Eur J Biochem. 1989;185:41–50.
  • Chen TG, Lee JJ, Lin KH, et al. Antiplatelet activity of caffeic acid phenethyl ester is mediated through a cyclic GMP-dependent pathway in human platelets. Chin J Physiol. 2007;50(3):121–126.
  • Wang X, Pang J, Maffucci JA, et al. Pharmacokinetics of caffeic acid phenethyl ester and its catechol-ring fluorinated derivative following intravenous administration to rats. Biopharm Drug Dispos. 2009;30(5):221–228.
  • Catalgo BS, Batirel Y, Taga NK. Ozer Resveratrol: French paradox revisited Front. Pharmacol. 2012;3:141.
  • Göçmen AY, Burgucu D, Gümüşlü S. Effect of resveratrol on platelet activation in hypercholesterolemic rats: CD40-CD40L system as a potential target. Appl Physiol Nutr Metab. 2011;36(3):323–330.
  • Malinowska J, Olas B. Response of blood platelets to resveratrol during a model of hyperhomocysteinemia. Platelets. 2011;22(4):277–283.
  • Yang Y, Wang X, Zhang L, et al. Inhibitory effects of resveratrol on platelet activation induced by thromboxane A(2) receptor agonist in human platelets. Am J Chin Med. 2011;39(1):145–159.
  • Yang YM, Wang XX, Wang SJ, et al. Suppressive effect in vitro of resveratrol on ADP induced human platelet aggregation and its active mechanism. Yao Xue Xue Bao. 2008;43(4):356–360.
  • Wu CC, Wu CI, Wang WY, et al. Low concentrations of resveratrol potentiates the antiplatelet effect of prostaglandins. Planta Med. 2007;73(5):439–443.
  • Stef G, Csiszar A, Lerea K, et al. Resveratrol inhibits aggregation of platelets from high-risk cardiac patients with aspirin resistance. J Cardiovasc Pharmacol. 2006;48(2):1–5.
  • Olas B, Nowak P, Ponczek M, et al. Resveratrol, a natural phenolic compound may reduce carbonylation proteins induced by peroxynitrite in blood platelets. Gen Physiol Biophys. 2006;25(2):215–222.
  • Olas B, Wachowicz B, Holmsen H, et al. Resveratrol inhibits polyphosphoinositide metabolism in activated platelets. Biochim Biophys Acta. 2005;1714(2):125–133.
  • Toliopoulos IK, Simos YV, Oikonomidis S, et al. Resveratrol diminishes platelet aggregation and increases susceptibility of K562 tumor cells to natural killer cells. Indian J Biochem Biophys. 2013;50(1):14–18.
  • Shen MY, Hsiao G, Liu CL, et al. Inhibitory mechanisms of resveratrol in platelet activation: pivotal roles of p38 MAPK and NO/cyclic GMP. Br J Haematol. 2007;139(3):475–485.
  • Liu J, Pestina TI, Berndt MC, et al. Botrocetin/VWF-induced signaling through GPIb-IX-V produces TxA2 in an alphaIIbbeta3–and aggregation-independent manner. Blood. 2005;106:2750–2756.
  • Nakamura F, Minshall RD, Le Breton GC, et al. Thromboxane A2 mediates the stimulation of inositol 1, 4, 5-trisphosphate production and intracellular calcium mobilization by bradykinin in neonatal rat ventricular cardiomyocytes. Hypertension. 1996;28:444–449.
  • Muzzio M, Huang Z, Hu SC, et al. Determination of resveratrol and its sulfate and glucuronide metabolites in plasma by LC-MS/MS and their pharmacokinetics in dogs. J Pharm Biomed Anal. 2012;59:201–208.
  • Chang CC, Lu WJ, Chiang CW, et al. Potent antiplatelet activity of sesamol in an in vitro and in vivo model: pivotal roles of cyclic AMP and p38 mitogen-activated protein kinase. J Nutr Biochem. 2010;21(12):1214–1221.
  • Chang CC, Lu WJ, Ong ET, et al. A novel role of sesamol in inhibiting NF-κB-mediated signaling in platelet activation. J Biomed Sci. 2011;14:93.
  • Thushara RM, Hemshekhar M, Sunitha K, et al. Sesamol induces apoptosis in human platelets via reactive oxygen species-mediated mitochondrial damage. Biochimie. 2013;95(11):2060–2068.
  • Jan KC, Ho CT, Hwang LS. Bioavailability and tissue distribution of sesamol in rat. J Agric Food Chem. 2008;56(16):7032–7037.
  • Thisoda P, Rangkadilok N, Pholphana N, et al. Inhibitory effect of Andrographis paniculata extract and its active diterpenoids on platelet aggregation. Eur J Pharmacol. 2006;553(1–3):39–45.
  • Lu WJ, Lee JJ, Chou DS, et al. A novel role of andrographolide, an NF-kappa B inhibitor, on inhibition of platelet activation: the pivotal mechanisms of endothelial nitric oxide synthase/cyclic GMP. J Mol Med. 2011;89:1261–1273.
  • Lu WJ, Lin KH, Hsu MJ, et al. Suppression of NF-kB signaling by andrographolide with a novel mechanism in human platelets: Regulatory roles of the p38 MAPK-hydroxyl radical-ERK2 cascade. Biochem Pharmacol. 2012;84:914–924.
  • Chen HW, Huang CS, Li CC, et al. Bioavailability of andrographolide and protection against carbon tetrachloride-induced oxidative damage in rats. Toxicol Appl Pharmacol. 2014;280(1):1–9.
  • Panossian A, Hovhannisyan A, Mamikonyan G, et al. Pharmacokinetic and oral bioavailability of andrographolide from Andrographis paniculata fixed combination Kan Jang in rats and human. Phytomedicine. 2000;7(5):351–364.
  • Chuang WY, Kung PH, Kuo CY, et al. Sulforaphane prevents human platelet aggregation through inhibiting the phosphatidylinositol 3-kinase/Akt pathway. Thromb Haemost. 2013;109(6):1120–1130.
  • Jayakumar T, Chen WF, Lu WJ, et al. A novel antithrombotic effect of sulforaphane via activation of platelet adenylate cyclase: ex vivo and in vivo studies. J Nutr Biochem. 2013;24(6):1086–1095.
  • Oh CH, Shin JI, Mo SJ, et al. Antiplatelet activity of L-sulforaphane by regulation of platelet activation factors, glycoprotein IIb/IIIa and thromboxane A2. Blood Coagul Fibrinolysis. 2013;24(5):498–504.
  • Lee YM, Hsieh KH, Lu WJ, et al. Xanthohumol, a Prenylated Flavonoid from Hops (Humuluslupulus), prevents platelet activation in human platelets. Evid Based Complement Alternat Med. 2012;2012:852362.
  • Krenn BM, Gaudernak E, Holzer B, et al. Antiviral activity of the zinc ionophores pyrithione and hinokitiol against picornavirus infections. J Virol. 2009;83(1):58–64.
  • Liu S, Yamauchi H. Hinokitiol a metal chelator derived from natural plants, suppresses cell growth and disrupts androgen receptor signaling in prostate carcinoma cell lines. Biochem Biophys Res Commun. 2006;351:26–32.
  • Lin KH, Kuo JR, Lu WJ, et al. Hinokitiol inhibits platelet activation ex vivo and thrombus formation in vivo. Biochem Pharmacol. 2013;85:1478–1485.
  • Lu WJ, Wu MP, Lin KH, et al. Hinokitiol is a novel glycoprotein VI antagonist on human platelets. Platelets. 2014;25(8):595–602.
  • Cho YM, Hasumura M, Takami S, et al. A 13-week subchronic toxicity study of hinokitiol administered in the diet to F344 rats. Food Chem Toxicol. 2011;49(8):1782–1786.
  • Lu WJ, Chang NC, Jayakumar T, et al. Ex vivo and in vivo studies of CME-1, a novel polysaccharide purified from the mycelia of Cordyceps sinensis that inhibits human platelet activation by activating adenylate cyclase/cyclic AMP. Thromb Res. 2014;134(6):1301–1310.
  • Chang Y, Hsu WH, Lu WJ, et al. Inhibitory mechanisms of CME-1, a novel polysaccharide from the mycelia of Cordyceps sinensis, in platelet activation. Curr Pharm Biotechnol. 2015;16(5):451–461.
  • Sugiura T1, Nakamura H. Metallothionein in platelets. Int Arch Allergy Immunol. 1994;103(4):341–348.
  • Sheu JR, Hsiao G, Shen MY, et al. Inhibitory mechanisms of metallothionein on platelet aggregation in in vitro and platelet plug formation in in vivo experiments. Exp Biol Med. 2003;228:1321–1328.
  • Penkowa M, Hidalgo J. Metallothionein I+II expression and their role in experimental autoimmune encephalomyelitis. Glia. 2000;32(3):247–263.
  • Lee YM, Lee JJ, Shen MY, et al. Inhibitory mechanisms of activated matrix metalloproteinase-9 on platelet activation. Eur J Pharmacol. 2006;537:52–58.
  • Kaushansky K. The molecular mechanisms that control thrombopoiesis. J Clin Invest. 2005;115:3339–3347.
  • Lane WJ, Dias S, Hattori K, et al. Stromal-derived factor 1-induced megakaryocyte migration and platelet production is dependent on matrix metalloproteinases. Blood. 2000;96:4152–4159.
  • Sheu JR, Fong TH, Liu CM, et al. Expression of matrix metalloproteinase-9 in human platelets: regulation of platelet activation in in vitro and in vivo studies. Br J Pharmacol. 2004;143(1):193–201.
  • Lee YM, Lee JJ, Shen MY, et al. Inhibitory mechanisms of activated matrix metalloproteinase-9 on platelet activation. Eur J Pharmacol. 2006;537(1–3):52–58.
  • Papp J, Kenyeres P, Toth K. Clinical importance of antiplatelet drugs in cardiovascular diseases. Clin Hemorheol Microcirc. 2013;53(1–2):81–96.
  • Dobrydneva Y, Weatherman RV, Trebley JP, et al. Tamoxifen stimulates calcium entry into human platelets. J Cardiovasc Pharmacol. 2007;50(4):380–390.
  • Chang Y, Lee JJ, Chen WF, et al. A novel role for tamoxifen in the inhibition of human platelets. Transl Res. 2011;157(2):81–91.
  • Nayak L, Schmaier AH. A platelet acquired storage pool disorder associated with Tamoxifen therapy. Case Rep Hematol. 2012;2012:1–3.
  • Ramström AS, Fagerberg IH, Lindahl TL. A flow cytometric assay for the study of dense granule storage and release in human platelets. Platelets. 1999;10:153–158.
  • Zhou L, Schmaier AH. Platelet aggregation testing in platelet-rich plasma: description of procedures with the aim to develop standards in the field. Am J Clin Pathol. 2005;123:172–183.
  • Nanetti L, Camilletti A, Francucci CM, et al. Role of raloxifene on platelet metabolism and plasma lipids. Eur J Clin Invest. 2008;38:117–125.
  • Decensi A, Gandini S, Guerrieri-Gonzaga A, et al. Effect of blood tamoxifen concentrations on surrogate biomarkers in a trial of dose reduction in healthy women. J Clin Oncol. 1999;17:2633–2638.
  • Decensi A, Robertson C, Rotmensz N, et al. Italian Chemoprevention Group. Effect of tamoxifen and transdermal hormone replacement therapy on cardiovascular risk factors in a prevention trial. Br J Cancer. 1998;78:572–578.
  • Love RR, Wiebe DA, Feyzi JM, et al. Effects of tamoxifen on cardiovascular risk factors in postmenopausal women after 5 years of treatment. J Natl Cancer Inst. 1994;86:1534–1539.
  • Bonetti PO, Lerman LO, Napoli C, et al. Statin effects beyond lipid lowering-are they clinically relevant? Eur Heart J. 2003;24:225–248.
  • Ruggeri ZM. Platelets in atherothrombosis. Nat Med. 2002;8:1227–1234.
  • Gaddam V, Li DY, Mehta JL. Anti-thrombotic effects of atorvastatin-an effect unrelated to lipid lowering. J Cardiovasc Pharmacol Ther. 2002;7:247–253.
  • Ma LP, Nie DN, Hsu SX, et al. Inhibition of platelet aggregation and expression of alpha granule membrane protein 140 and thromboxane B2 with pravastatin therapy for hypercholesterolemia. J Assoc Acad Minor Phys. 2002;13:23–26.
  • Ali FY, Armstrong PC, Dhanji AR, et al. Antiplatelet actions of statins and fibrates are mediated by PPARs. Arterioscler Thromb Vasc Biol. 2009;29:706–711.
  • Moscardó A, Vallés J, Latorre A, et al. Reduction of platelet cytosolic phospholipase A2 activity by atorvastatin and simvastatin: biochemical regulatory mechanisms. Thromb Res. 2013;131(4):e154–159.
  • Lee YM, Chen WF, Chou DS, et al. Cyclic nucleotides and mitogen-activated protein kinases: regulation of simvastatin in platelet activation. J Biomed Sci. 2010;17:45.
  • Jayachandran M, Miller VM. Human platelets contain estrogen receptor α, caveolin-1 and estrogen receptor associated proteins. Platelets. 2003;14:75–81.
  • Akarasereenont P, Tripatara P, Chotewuttakorn S, et al. The effects of estrone, estradiol and estriol on platelet aggregation induced by adrenaline and adenosine diphosphate. Platelets. 2006;17(7):441–447.
  • Bar J, Lahav J, Hod M, et al. Regulation of platelet aggregation and adenosine triphosphate release in vitro by 17β-estradiol and medroxyprogesterone acetate in postmenopausal women. Thromb Haemost. 2000;84:695–700.
  • Chen L, Liu Y, Cui B, et al. 17β-estradiol partially attenuates the inhibition of nitric oxide synthase-3 byadvanced glycation end-products in human platelets. Clin Exp Pharmacol Physiol. 2007;34:972–978.
  • Zerr-Fouineau M, Jourdain M, Boesch C, et al. Certain progestins prevent the enhancing effect of 17β-estradiol on NO mediated inhibition of platelet aggregation by endothelial cells. Arterioscler Thromb Vasc Biol. 2009;29:586–593.
  • Moro L, Reineri S, Piranda D, et al. Nongenomic effects of 17 (beta)-estradiol in human platelets: potentiation of thrombin-induced aggregation through estrogen receptor (beta) and Src kinase. Blood. 2005;105:115–121.
  • Nakano Y, Oshima T, Matsuura H, et al. Effect of 17β- estradiol on inhibition of platelet aggregation in vitro is mediated by an increase in NO synthesis. Arterioscler Thromb Vasc Biol. 1998;18:961–967.
  • Lu WJ, Lee JJ, Chou DS, et al. Inhibitory signaling of 17β-estradiol in platelet activation: the pivotal role of cyclic AMP-mediated nitric oxide synthase activation. Eur J Pharmacol. 2010;649:140–149.
  • Volterrani M, Rosano G, Coats A, et al. Estrogen acutely increases peripheral blood flow in postmenopausal women. Am J Med. 1995;99:119–122.
  • Vitale C, Mercuro G, Cerquetani E, et al. Time since menopause influences the acute and chronic effect of estrogens on endothelial function. Arterioscler Thromb Vasc Biol. 2008;28:348–352.
  • Haas E, Meyer MR, Schurr U, et al. Differential effects of 17beta-estradiol on function and expression of estrogen receptor alpha, estrogen receptor beta, and GPR30 in arteries and veins of patients with atherosclerosis. Hypertension. 2007;49:1358–1363.
  • Chen ZM, Jiang LX, Chen YP, et al. Addition of clopid-ogrel to aspirin in 45,852 patients with acute myocar-dial infarction: randomised placebo-controlled trial. Lancet. 2005;366:1607–1621.
  • Herbert JM, Savi P. P2Y12, a new platelet ADP receptor, target of clopidogrel. SeminVasc Med. 2003;3:113–122.
  • Sofi F, Marcucci R, Gori AM, et al. Clopidogrel non-responsiveness and risk of cardiovascular morbidity. An updated meta-analysis. Thromb Haemost. 2010;103:841–848.
  • Brandt JT, Payne CD, Wiviott SD, et al. A comparison of pra-sugrel and clopidogrel loading doses on platelet function: magnitude of platelet inhibition is related to active metabolite formation. Am Heart J. 2007;153:9–16.
  • Montalescot G, Sideris G, Cohen R, et al. Prasugrel compared with high-dose clopidogrel in acute coronary syndrome. The randomised, double-blind ACAPULCO study. Thromb Haemost. 2010;103:213–223.
  • Angiolillo DJ, Badimon JJ, Saucedo JF, et al. A pharmacodynamic comparison of prasugrel vs. high dose clopidogrel in patients with type 2 diabetes mellitus and coronary artery disease: results of the optimizing anti-platelet therapy in diabetes mellitus (OPTIMUS)-3 Trial. Eur Heart J. 2011;32:838–846.
  • Wiviott SD, Trenk D, Frelinger AL, et al. Prasugrel com-pared with high loading- and maintenance-dose clopidogrel inpatients with planned percutaneous coronary intervention: the prasugrel in comparison to clopidogrel for inhibition of platelet activation and aggregation-thrombolysis in myocardial infarction 44 trial. Circulation. 2007;116:2923–2932.
  • Wallentin L. P2Y(12) inhibitors: differences in properties and mechanisms of action and potential consequences for clinical use. Eur Heart J. 2009;30:1964–1977.
  • Storey RF, Angiolillo DJ, Patil SB, et al. Inhibitory effects of ticagrelor compared with clopidogrel on platelet function in patients with acute coronary syndromes: the PLATO (Platelet inhibition and patient outcomes) PLATELET substudy. J Am Coll Cardiol. 2010;56:1456–1462.
  • Gurbel PA, Bliden KP, Butler K, et al. Response to ticagrelor in clopidogrel non responders and responders and effect of switching therapies: the RESPOND study. Circulation. 2010;121:1188–1199.
  • Steiner S, Seidinger D, Huber K, et al. Effect of glycoprotein IIb/IIIa antagonist abciximab on monocyte-platelet aggregates and tissue factor expression. Arterioscler Thromb Vasc Biol. 2003;23:1697–1702.
  • Ibbotson T, McGavin JK, Goa KL. Abciximab: an updated review of its therapeutic use in patients with ischaemic heart disease undergoing percutaneous coronary revascularisation. Drugs. 2003;63:1121–1163.
  • Braunwald’s heart disease: a textbook of cardiovascular medicine. 9th ed. Philadelphia: Elsevier Saunders; 2012.
  • Hamilton J. Protease-activated receptors as targets for antiplatelet therapy. Blood Rev. 2009;23:61–65.
  • Kakkos SK, Nicolaides AN. S-18886 Servier. Curr Opin Investig Drugs. 2002;3:1324–1327.
  • Shalito I, Kopyleva O, Serebruany V. Novel antiplatelet agents in development: prasugrel, ticagrelor, and cangrelor and beyond. Am J Ther. 2009;16:451–458.

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.